1
|
Mazurkiewicz M, Bodnar P, Blachut D, Chwalba T, Wagner W, Barczyk E, Romuk E, Jacheć W, Wojciechowska C. Adipokines and Adipose Tissue: The Role and Use of Sodium-Glucose Co-Transporter-2 (SGLT-2) Inhibitors in Patients with Diabetes or Heart Failure. Biomedicines 2025; 13:1098. [PMID: 40426925 PMCID: PMC12108695 DOI: 10.3390/biomedicines13051098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Sodium-glucose co-transporter-2 (SGLT-2) inhibitors have become integral in treating both diabetes mellitus and heart failure, independent of left ventricular ejection fraction. Their pleiotropic effect influences multiple mechanisms, enhancing the function of various systems within the body. They exhibit nephroprotective and cardioprotective effects by improving cell metabolism, endothelial function, and slowing the fibrosis of the cardiac muscle, and they also have a beneficial impact on other organs. At the cellular level, they protect against the harmful effects of free radicals both by lowering glucose levels and by supporting the function of the antioxidant system. Moreover, SGLT-2 inhibitors can modify the metabolism of adipocytes by affecting the production of cytokines such as adiponectin-which increases insulin sensitivity, leading to weight loss and improved glycemic control.
Collapse
Affiliation(s)
- Michalina Mazurkiewicz
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Patryk Bodnar
- Department of Anaesthesiology and Intensive Care, Clinical Hospital in Czeladź, Szpitalna 40 Street, 41-250 Czeladź, Poland;
| | - Dominika Blachut
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Tomasz Chwalba
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Wiktor Wagner
- Student Research Team at the Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (W.W.); (E.B.)
| | - Eliza Barczyk
- Student Research Team at the Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (W.W.); (E.B.)
| | - Ewa Romuk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Street, 41-808 Zabrze, Poland;
| | - Wojciech Jacheć
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| | - Celina Wojciechowska
- Second Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Specialistic Hospital in Zabrze, M. C. Skłodowskiej 10 Street, 41-800 Zabrze, Poland; (D.B.); (T.C.); (W.J.); (C.W.)
| |
Collapse
|
2
|
Conde KM, Wong H, Fang S, Li Y, Yu M, Deng Y, Liu Q, Fang X, Wang M, Shi Y, Ginnard OZ, Yang Y, Tu L, Liu H, Liu H, Yin N, Bean JC, Han J, Burt ME, Jossy SV, Yang Y, Tong Q, Arenkiel BR, Wang C, He Y, Xu Y. Serotonin neurons integrate GABA and dopamine inputs to regulate meal initiation. Metabolism 2025; 163:156099. [PMID: 39667432 PMCID: PMC11924950 DOI: 10.1016/j.metabol.2024.156099] [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: 10/10/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
Obesity is a growing global health epidemic with limited orally administered therapeutics. Serotonin (5-HT) is one neurotransmitter which remains an excellent target for new weight-loss therapies, but a gap remains in understanding the mechanisms involved in 5-HT produced in the dorsal Raphe nucleus (DRN) and its involvement in meal initiation. Using an optogenetic feeding paradigm, we showed that the 5-HTDRN➔arcuate nucleus (ARH) circuit plays a role in meal initiation. Incorporating electrophysiology and ChannelRhodopsin-2-Assisted Circuit Mapping, we demonstrated that 5-HTDRN neurons receive inhibitory input partially from GABAergic neurons in the DRN, and the 5-HT response can be enhanced by hunger. Additionally, deletion of the GABAA receptor subunit in 5-HT neurons inhibits meal initiation with no effect on the satiation process. Finally, we identified the role of dopaminergic inputs via dopamine receptor D2 in enhancing the response to GABA-induced feeding. Thus, our results indicate that 5-HTDRN neurons are inhibited by synergistic inhibitory actions of GABA and dopamine, for the initiation of a meal.
Collapse
Affiliation(s)
- Kristine M Conde
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - HueyZhong Wong
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shuzheng Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongxiang Li
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Meng Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yue Deng
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingzhuo Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xing Fang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuhan Shi
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Olivia Z Ginnard
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuxue Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hesong Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Na Yin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jonathan C Bean
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Junying Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Megan E Burt
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sanika V Jossy
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chunmei Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yang He
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
3
|
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
|
4
|
Stark R, Dempsey H, Kleeman E, Sassi M, Osborne-Lawrence S, Sheybani-Deloui S, Rushby HJ, Mirth CK, Austin-Muttitt K, Mullins J, Zigman JM, Davies JS, Andrews ZB. Hunger signalling in the olfactory bulb primes exploration, food-seeking and peripheral metabolism. Mol Metab 2024; 89:102025. [PMID: 39236785 PMCID: PMC11471258 DOI: 10.1016/j.molmet.2024.102025] [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: 07/09/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
OBJECTIVE Although the metabolic state of an organism affects olfactory function, the precise mechanisms and their impact on behavior and metabolism remain unknown. Here, we assess whether ghrelin receptors (GHSRs) in the olfactory bulb (OB) increase olfactory function and influence foraging behaviors and metabolism. METHODS We performed a detailed behavioural and metabolic analysis in mice lacking GHSRs in the OB (OBGHSR deletion). We also analsyed OB scRNA-seq and spatial transcriptomic datasets to assess GHSR+ cells in the main and accessory olfactory bulbs, as well as the anterior olfactory nucleus. RESULTS OBGHSR deletion affected olfactory discrimination and habituation to both food and non-food odors. Anxiety-like and depression-like behaviors were significantly greater after OBGHSR deletion, whereas exploratory behavior was reduced, with the greatest effect under fasted conditions. OBGHSR deletion impacted feeding behavior as evidenced by altered bout number and duration, as well as buried food-seeking. OBGHSR deletion increased body weight and fat mass, spared fat utilisation on a chow diet and impaired glucose metabolism indicating metabolic dysfunction. Cross referenced analysis of OB scRNA-seq and spatial transcriptomic datasets revealed GHSR+ glutamate neurons in the main and accessory olfactory bulbs, as well as the anterior olfactory nucleus. Ablation of glutamate neurons in the OB reduced ghrelin-induced food finding and phenocopied results seen after OBGHSR deletion. CONCLUSIONS OBGHSRs help to maintain olfactory function, particularly during hunger, and facilitate behavioral adaptations that optimise food-seeking in anxiogenic environments, priming metabolic pathways in preparation for food consumption.
Collapse
Affiliation(s)
- Romana Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Harry Dempsey
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Elizabeth Kleeman
- The Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, Australia
| | - Martina Sassi
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sepideh Sheybani-Deloui
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Helen J Rushby
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Christen K Mirth
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Karl Austin-Muttitt
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Jonathan Mullins
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey S Davies
- Institute of Life Sciences, School of Medicine, Swansea University, Swansea, UK
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
5
|
Han HW, Pradhan G, Villarreal D, Kim DM, Jain A, Gaharwar A, Tian Y, Guo S, Sun Y. GHSR Deletion in β-Cells of Male Mice: Ineffective in Obesity, but Effective in Protecting against Streptozotocin-Induced β-Cell Injury in Aging. Nutrients 2024; 16:1464. [PMID: 38794702 PMCID: PMC11123813 DOI: 10.3390/nu16101464] [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: 04/15/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Insulin secretion from pancreatic β cells is a key pillar of glucose homeostasis, which is impaired under obesity and aging. Growth hormone secretagogue receptor (GHSR) is the receptor of nutrient-sensing hormone ghrelin. Previously, we showed that β-cell GHSR regulated glucose-stimulated insulin secretion (GSIS) in young mice. In the current study, we further investigated the effects of GHSR on insulin secretion in male mice under diet-induced obesity (DIO) and streptozotocin (STZ)-induced β-cell injury in aging. β-cell-specific-Ghsr-deficient (Ghsr-βKO) mice exhibited no glycemic phenotype under DIO but showed significantly improved ex vivo GSIS in aging. We also detected reduced insulin sensitivity and impaired insulin secretion during aging both in vivo and ex vivo. Accordingly, there were age-related alterations in expression of glucose transporter, insulin signaling pathway, and inflammatory genes. To further determine whether GHSR deficiency affected β-cell susceptibility to acute injury, young, middle-aged, and old Ghsr-βKO mice were subjected to STZ. We found that middle-aged and old Ghsr-βKO mice were protected from STZ-induced hyperglycemia and impaired insulin secretion, correlated with increased expression of insulin signaling regulators but decreased pro-inflammatory cytokines in pancreatic islets. Collectively, our findings indicate that β-cell GHSR has a major impact on insulin secretion in aging but not obesity, and GHSR deficiency protects against STZ-induced β-cell injury in aging.
Collapse
Affiliation(s)
- Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Geetali Pradhan
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniel Villarreal
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Akhilesh Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Yanan Tian
- Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.W.H.)
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Mahalingam S, Bellamkonda R, Kharbanda KK, Arumugam MK, Kumar V, Casey CA, Leggio L, Rasineni K. Role of ghrelin hormone in the development of alcohol-associated liver disease. Biomed Pharmacother 2024; 174:116595. [PMID: 38640709 PMCID: PMC11161137 DOI: 10.1016/j.biopha.2024.116595] [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: 01/25/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
Fatty liver is the earliest response of the liver to excessive alcohol consumption. Previously we identified that chronic alcohol administration increases levels of stomach-derived hormone, ghrelin, which by reducing circulating insulin levels, ultimately contributes to the development of alcohol-associated liver disease (ALD). In addition, ghrelin directly promotes fat accumulation in hepatocytes by enhancing de novo lipogenesis. Other than promoting ALD, ghrelin is known to increase alcohol craving and intake. In this study, we used a ghrelin receptor (GHSR) knockout (KO) rat model to characterize the specific contribution of ghrelin in the development of ALD with emphasis on energy homeostasis. Male Wistar wild type (WT) and GHSR-KO rats were pair-fed the Lieber-DeCarli control or ethanol diet for 6 weeks. At the end of the feeding period, glucose tolerance test was conducted, and tissue samples were collected. We observed reduced alcohol intake by GHSR-KOs compared to a previous study where WT rats were fed ethanol diet ad libitum. Further, when the WTs were pair-fed to GHSR-KOs, the KO rats exhibited resistance to develop ALD through improving insulin secretion/sensitivity to reduce adipose lipolysis and hepatic fatty acid uptake/synthesis and increase fatty acid oxidation. Furthermore, proteomic data revealed that ethanol-fed KO exhibit less alcohol-induced mitochondrial dysfunction and oxidative stress than WT rats. Proteomic data also confirmed that the ethanol-fed KOs are insulin sensitive and are resistant to hepatic steatosis development compared to WT rats. Together, these data confirm that inhibiting ghrelin action prevent alcohol-induced liver and adipose dysfunction independent of reducing alcohol intake.
Collapse
Affiliation(s)
- Sundararajan Mahalingam
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ramesh Bellamkonda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Madan Kumar Arumugam
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vikas Kumar
- Mass Spectrometry and Proteomic Core Facility, University of Nebraska Medical Center, Omaha, NE, USA; Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Carol A Casey
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Bethesda, Baltimore, MD, USA; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA; Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
7
|
Mitra A, Mandal S, Bose B, Shenoy P S. Unlocking the Potential of Obestatin: A Novel Peptide Intervention for Skeletal Muscle Regeneration and Prevention of Atrophy. Mol Biotechnol 2024; 66:948-959. [PMID: 38198052 DOI: 10.1007/s12033-023-01011-7] [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: 10/01/2023] [Accepted: 11/27/2023] [Indexed: 01/11/2024]
Abstract
Obestatin is derived from the same gene as that of ghrelin and their functions were perceived to be antagonistic. Recent developments have shown that although they are known to have contradictory functions, effect of obestatin on skeletal muscle regeneration is similar to that of ghrelin. Obestatin works through a receptor called GPR39, a ghrelin and motilin family receptor and transduces signals in skeletal muscle similar to that of ghrelin. Not only there is a similarity in the receptor family, but also obestatin targets similar proteins and transcription factors as that of ghrelin (for example, FoxO family members) for salvaging skeletal muscle atrophy. Moreover, like ghrelin, obestatin also works by inducing the transcription of Pax7 which is required for muscle stem cell mobilisation. Hence, there are quite some evidences which points to the fact that obestatin can be purposed as a peptide intervention to prevent skeletal muscle wasting and induce myogenesis. This review elaborates these aspects of obestatin which can be further exploited and addressed to bring obestatin as a clinical intervention towards preventing skeletal muscle atrophy and sarcopenia.
Collapse
Affiliation(s)
- Akash Mitra
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, Karnataka, 575018, India
| | - Samanwita Mandal
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, Karnataka, 575018, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, Karnataka, 575018, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
8
|
Carneiro L, Fenech C, Liénard F, Grall S, Abed B, Haydar J, Allard C, Desmoulins L, Paccoud R, Brindisi MC, Mouillot T, Brondel L, Fioramonti X, Pénicaud L, Jacquin-Piques A, Leloup C. Hypothalamic Glucose Hypersensitivity-Induced Insulin Secretion in the Obese Zücker Rat Is Reversed by Central Ghrelin Treatment. Antioxid Redox Signal 2024; 40:837-849. [PMID: 36656675 DOI: 10.1089/ars.2022.0031] [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] [Indexed: 01/20/2023]
Abstract
Aims: Part of hypothalamic (mediobasal hypothalamus [MBH]) neurons detect changes in blood glucose levels that in turn coordinate the vagal control of insulin secretion. This control cascade requires the production of mitochondrial reactive oxygen species (mROS), which is altered in models of obesity and insulin resistance. Obese, insulin-resistant Zücker rats are characterized by hypothalamic hypersensitivity to glucose. This initiates an abnormal vagus-induced insulin secretion, associated with an overproduction of mROS in response to a low glucose dose. Here, we hypothesized that ghrelin, known to buffer reactive oxygen species (ROS) via mitochondrial function, may be a major component of the hypothalamic glucose hypersensitivity in the hypoghrelinemic obese Zücker rat. Results: Hypothalamic glucose hypersensitivity-induced insulin secretion of Zücker obese rats was reversed by ghrelin pretreatment. The overproduction of MBH mROS in response to a low glucose load no longer occurred in obese rats that had previously received the cerebral ghrelin infusion. This decrease in mROS production was accompanied by a normalization of oxidative phosphorylation (OXPHOS). Conversely, blocking the action of ghrelin with a growth hormone secretagogue receptor antagonist in a model of hyperghrelinemia (fasted rats) completely restored hypothalamic glucose sensing-induced insulin secretion that was almost absent in this physiological situation. Accordingly, ROS signaling and mitochondrial activity were increased by the ghrelin receptor antagonist. Innovation: These results demonstrate for the first time that ghrelin addressed only to the brain could have a protective effect on the defective control of insulin secretion in the insulin-resistant, hypoghrelinemic obese subject. Conclusions: Ghrelin, through its action on OXPHOS, modulates mROS signaling in response to cerebral hyperglycemia and the consequent vagal control of insulin secretion. In insulin-resistant obese states, brain hypoghrelinemia could be responsible for the nervous defect in insulin secretion.
Collapse
Affiliation(s)
- Lionel Carneiro
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Toulouse, France
| | - Claire Fenech
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Fabienne Liénard
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Sylvie Grall
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Besma Abed
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Joulia Haydar
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Camille Allard
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
- University of Bordeaux, INSERM U1215, Neurocentre Magendie, Bordeaux, France
| | - Lucie Desmoulins
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Romain Paccoud
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Marie-Claude Brindisi
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Thomas Mouillot
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Laurent Brondel
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Xavier Fioramonti
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
- NutriNeuro, UMR 1286 INRAE, Bordeaux University, Bordeaux INP, Neurocampus, Bordeaux, France
| | - Luc Pénicaud
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
- STROMALab, CNRS ERL 5311, Toulouse, France
| | - Agnès Jacquin-Piques
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| | - Corinne Leloup
- Centre des Sciences du Goût et de l'Alimentation, UMR Université de Bourgogne, CNRS 6265, INRAE 1324, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
9
|
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
|
10
|
Wee RWS, Mishchanchuk K, AlSubaie R, Church TW, Gold MG, MacAskill AF. Internal-state-dependent control of feeding behavior via hippocampal ghrelin signaling. Neuron 2024; 112:288-305.e7. [PMID: 37977151 DOI: 10.1016/j.neuron.2023.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Hunger is an internal state that not only invigorates feeding but also acts as a contextual cue for higher-order control of anticipatory feeding-related behavior. The ventral hippocampus is crucial for differentiating optimal behavior across contexts, but how internal contexts such as hunger influence hippocampal circuitry is unknown. In this study, we investigated the role of the ventral hippocampus during feeding behavior across different states of hunger in mice. We found that activity of a unique subpopulation of neurons that project to the nucleus accumbens (vS-NAc neurons) increased when animals investigated food, and this activity inhibited the transition to begin eating. Increases in the level of the peripheral hunger hormone ghrelin reduced vS-NAc activity during this anticipatory phase of feeding via ghrelin-receptor-dependent increases in postsynaptic inhibition and promoted the initiation of eating. Together, these experiments define a ghrelin-sensitive hippocampal circuit that informs the decision to eat based on internal state.
Collapse
Affiliation(s)
- Ryan W S Wee
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Karyna Mishchanchuk
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Rawan AlSubaie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Timothy W Church
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Matthew G Gold
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK
| | - Andrew F MacAskill
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, UK.
| |
Collapse
|
11
|
Kim DM, Lee JH, Pan Q, Han HW, Shen Z, Eshghjoo S, Wu CS, Yang W, Noh JY, Threadgill DW, Guo S, Wright G, Alaniz R, Sun Y. Nutrient-sensing growth hormone secretagogue receptor in macrophage programming and meta-inflammation. Mol Metab 2024; 79:101852. [PMID: 38092245 PMCID: PMC10772824 DOI: 10.1016/j.molmet.2023.101852] [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: 10/11/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVE Obesity-associated chronic inflammation, aka meta-inflammation, is a key pathogenic driver for obesity-associated comorbidity. Growth hormone secretagogue receptor (GHSR) is known to mediate the effects of nutrient-sensing hormone ghrelin in food intake and fat deposition. We previously reported that global Ghsr ablation protects against diet-induced inflammation and insulin resistance, but the site(s) of action and mechanism are unknown. Macrophages are key drivers of meta-inflammation. To unravel the role of GHSR in macrophages, we generated myeloid-specific Ghsr knockout mice (LysM-Cre;Ghsrf/f). METHODS LysM-Cre;Ghsrf/f and control Ghsrf/f mice were subjected to 5 months of high-fat diet (HFD) feeding to induce obesity. In vivo, metabolic profiling of food intake, physical activity, and energy expenditure, as well as glucose and insulin tolerance tests (GTT and ITT) were performed. At termination, peritoneal macrophages (PMs), epididymal white adipose tissue (eWAT), and liver were analyzed by flow cytometry and histology. For ex vivo studies, bone marrow-derived macrophages (BMDMs) were generated from the mice and treated with palmitic acid (PA) or lipopolysaccharide (LPS). For in vitro studies, macrophage RAW264.7 cells with Ghsr overexpression or Insulin receptor substrate 2 (Irs2) knockdown were studied. RESULTS We found that Ghsr expression in PMs was increased under HFD feeding. In vivo, HFD-fed LysM-Cre;Ghsrf/f mice exhibited significantly attenuated systemic inflammation and insulin resistance without affecting food intake or body weight. Tissue analysis showed that HFD-fed LysM-Cre;Ghsrf/f mice have significantly decreased monocyte/macrophage infiltration, pro-inflammatory activation, and lipid accumulation, showing elevated lipid-associated macrophages (LAMs) in eWAT and liver. Ex vivo, Ghsr-deficient macrophages protected against PA- or LPS-induced pro-inflammatory polarization, showing reduced glycolysis, increased fatty acid oxidation, and decreased NF-κB nuclear translocation. At molecular level, GHSR metabolically programs macrophage polarization through PKA-CREB-IRS2-AKT2 signaling pathway. CONCLUSIONS These novel results demonstrate that macrophage GHSR plays a key role in the pathogenesis of meta-inflammation, and macrophage GHSR promotes macrophage infiltration and induces pro-inflammatory polarization. These exciting findings suggest that GHSR may serve as a novel immunotherapeutic target for the treatment of obesity and its associated comorbidity.
Collapse
Affiliation(s)
- Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Jong Han Lee
- Department of Marine Bioindustry, Hanseo University, Seosan 31962, South Korea; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Quan Pan
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Sahar Eshghjoo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Agilent technologies, Aanta Clara, CA 95051, USA
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA
| | - Wanbao Yang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - David W Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Genome Sciences and Society, Department of Cell Biology and Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Gus Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 77843, USA
| | - Robert Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA; Tlaloc Therapeutics Inc., College Station, TX 77845, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College Medicine, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Ibrahim M, Khalife L, Abdel-Latif R, Faour WH. Ghrelin hormone a new molecular modulator between obesity and glomerular damage. Mol Biol Rep 2023; 50:10525-10533. [PMID: 37924451 DOI: 10.1007/s11033-023-08866-8] [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: 06/30/2023] [Accepted: 09/27/2023] [Indexed: 11/06/2023]
Abstract
The incidence of glomerular diseases is increasing worldwide due to increased prevalence of obesity which is a major risk factor for type-2 diabetes mellitus and cardiovascular disorders.Ghrelin, an orexigenic peptide hormone, has been implicated in obesity, and its impact on the pathology and function of the kidneys was found to be significant. Ghrelin known to regulate energy homeostasis and growth hormone release, has been shown to modulate critical signaling pathways involved in the health and survival of podocytes. These derangements directly affect glomerular function and manifest as impaired glomerular filtration barrier and leakage of albumin into urine. Although the pathological features of the above-mentioned disorders are different, they interestingly lead to similar clinical features of glomerular damage. The pathological events are majorly initiated by endocrine imbalance leading to abnormal activation of downstream signaling pathways involved in the development of glomerulosclerosis. In fact, obesity increases the risk of developing chronic kidney disease by altering the secretion of pro-inflammatory cytokines and adipokines, activating the renin-angiotensin-aldosterone system (RAAS), promoting lipotoxicity, oxidative stress and fibrosis within the kidneys. Whilst these bioregulators are well described, their direct involvement in renal homeostasis is still mostly elusive. This review summarized previous and recent evidence on the endocrine properties of ghrelin and perivascular adipose tissue involved in modulating kidney physiology.
Collapse
Affiliation(s)
- Maroun Ibrahim
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Lynn Khalife
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Rania Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Miniya, Egypt
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon.
| |
Collapse
|
13
|
Alex Thomas M, Cui X, Artinian LR, Cao Q, Jing J, Silva FC, Wang S, Zigman JM, Sun Y, Shi H, Xue B. Crosstalk between Gut Sensory Ghrelin Signaling and Adipose Tissue Sympathetic Outflow Regulates Metabolic Homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.25.568689. [PMID: 38076894 PMCID: PMC10705268 DOI: 10.1101/2023.11.25.568689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The stomach-derived orexigenic hormone ghrelin is a key regulator of energy homeostasis and metabolism in humans. The ghrelin receptor, growth hormone secretagogue receptor 1a (GHSR), is widely expressed in the brain and gastrointestinal vagal sensory neurons, and neuronal GHSR knockout results in a profoundly beneficial metabolic profile and protects against diet-induced obesity (DIO) and insulin resistance. Here we show that in addition to the well characterized vagal GHSR, GHSR is robustly expressed in gastrointestinal sensory neurons emanating from spinal dorsal root ganglia. Remarkably, sensory neuron GHSR deletion attenuates DIO through increased energy expenditure and sympathetic outflow to adipose tissue independent of food intake. In addition, neuronal viral tract tracing reveals prominent crosstalk between gut non-vagal sensory afferents and adipose sympathetic outflow. Hence, these findings demonstrate a novel gut sensory ghrelin signaling pathway critical for maintaining energy homeostasis.
Collapse
Affiliation(s)
- M. Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA
| | - Xin Cui
- Department of Biology, Georgia State University, Atlanta, GA
| | | | - Qiang Cao
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jia Jing
- Department of Biology, Georgia State University, Atlanta, GA
| | - Felipe C. Silva
- Department of Biology, Georgia State University, Atlanta, GA
| | - Shirong Wang
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jeffrey M. Zigman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yuxiang Sun
- Department of Nutrition, Texas A & M University, College Station, TX
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA
| |
Collapse
|
14
|
Abhishek S, Deeksha W, Nethravathi KR, Davari MD, Rajakumara E. Allosteric crosstalk in modular proteins: Function fine-tuning and drug design. Comput Struct Biotechnol J 2023; 21:5003-5015. [PMID: 37867971 PMCID: PMC10589753 DOI: 10.1016/j.csbj.2023.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023] Open
Abstract
Modular proteins are regulatory proteins that carry out more than one function. These proteins upregulate or downregulate a biochemical cascade to establish homeostasis in cells. To switch the function or alter the efficiency (based on cellular needs), these proteins require different facilitators that bind to a site different from the catalytic (active/orthosteric) site, aka 'allosteric site', and fine-tune their function. These facilitators (or effectors) are allosteric modulators. In this Review, we have discussed the allostery, characterized them based on their mechanisms, and discussed how allostery plays an important role in the activity modulation and function fine-tuning of proteins. Recently there is an emergence in the discovery of allosteric drugs. We have also emphasized the role, significance, and future of allostery in therapeutic applications.
Collapse
Affiliation(s)
- Suman Abhishek
- Macromolecular Structural Biology lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Telangana 502284, India
| | - Waghela Deeksha
- Macromolecular Structural Biology lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Telangana 502284, India
| | | | - Mehdi D. Davari
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle 06120, Germany
| | - Eerappa Rajakumara
- Macromolecular Structural Biology lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Telangana 502284, India
| |
Collapse
|
15
|
Zhang H, Yan X, Lin A, Xia P, Su Y. Inhibition of ghrelin activity by the receptor antagonist [D-Lys3]-GHRP-6 enhances hepatic fatty acid oxidation and gluconeogenesis in a growing pig model. Peptides 2023; 166:171041. [PMID: 37301480 DOI: 10.1016/j.peptides.2023.171041] [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: 03/28/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
Despite its central role in regulating energy intake and metabolism, ghrelin is little understood when it comes to its effects on hepatic lipid and glucose metabolism. Growing pigs were intravenously injected with ghrelin receptor antagonist [D-Lys3]-GHRP-6 (DLys; 6 mg/kg body weight) for seven days to determine whether ghrelin plays a role in glucose and lipid metabolism. DLys treatment significantly reduced body weight gain and adipose histopathology found that DLys treatment dramatically reduced adipocyte size. DLys treatment significantly increased serum NEFA and insulin levels, hepatic glucose level and HOMA-IR, and significantly decreased serum TBA level of growing pigs after fasting. Moreover, DLys treatment changed the dynamics of serum metabolic parameters, including glucose, NEFA, TBA, insulin, GH, leptin, and cortisol. Liver transcriptome showed that DLys treatment affected the metabolism-related pathways. Compared with the control group, adipose tissue lipolysis (the adipose triglyceride lipase level was significantly increased), hepatic gluconeogenesis (the G6PC protein level was significantly increased) and fatty acid oxidation (the CPT1A protein level was significantly increased) were promoted in the DLys group. DLys treatment expanded degrees of oxidative phosphorylation in the liver, coming about in a higher NAD+ /NADH proportion and enactment of the SIRT1 signaling pathway. Additionally, the liver protein levels of the DLys group were significantly higher than those of the control group for GHSR, PPAR alpha, and PGC-1. To summarize, inhibition of ghrelin activity can significantly affect metabolism and alter energy levels by enhancing fat mobilization, hepatic fatty acid oxidation and gluconeogenesis without affecting fatty acid uptake and synthesis in the liver.
Collapse
Affiliation(s)
- He Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, 210095 Nanjing, China; College of Life Sciences, Xuzhou Medical University, 221004 Xuzhou, China
| | - Xiaoxi Yan
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, 210095 Nanjing, China
| | - Ailian Lin
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, 210095 Nanjing, China
| | - Pengke Xia
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, 210095 Nanjing, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, 210095 Nanjing, China.
| |
Collapse
|
16
|
Iwakura H, Ensho T, Ueda Y. Desacyl-ghrelin, not just an inactive form of ghrelin?-A review of current knowledge on the biological actions of desacyl-ghrelin. Peptides 2023:171050. [PMID: 37392995 DOI: 10.1016/j.peptides.2023.171050] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Desacyl-ghrelin is a form of ghrelin which lacks acyl-modification of the third serine residue of ghrelin. Originally, desacyl-ghrelin was considered to be just an inactive form of ghrelin. More recently, however, it has been suggested to have various biological activities, including control of food intake, growth hormone, glucose metabolism, and gastric movement, and is involved in cell survival. In this review, we summarize the current knowledge of the biological actions of desacyl-ghrelin and the proposed mechanisms by which it exerts the effects.
Collapse
Affiliation(s)
- Hiroshi Iwakura
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, 25-1 Shichibancho, Wakayama 640-8156, Japan.
| | - Takuya Ensho
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, 25-1 Shichibancho, Wakayama 640-8156, Japan
| | - Yoko Ueda
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, 25-1 Shichibancho, Wakayama 640-8156, Japan
| |
Collapse
|
17
|
Li N, Li N, Yang L, Gu H, Ji J, Zhou H, Zhu Q, Yu M, Sun Y, Zhou Y. GHSR1a deficiency suppresses inhibitory drive on dCA1 pyramidal neurons and contributes to memory reinforcement. Cereb Cortex 2023; 33:2612-2625. [PMID: 35797708 DOI: 10.1093/cercor/bhac230] [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: 04/08/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/12/2022] Open
Abstract
Growth hormone secretagogue receptor 1a (GHSR1a)-the receptor for orexigenic hormone ghrelin-is a G protein-coupled receptor that is widely distributed in the brain, including the hippocampus. Studies have demonstrated that genetic deletion of GHSR1a affects memory, suggesting the importance of ghrelin/GHSR1a signaling in cognitive control. However, current reports are controversial, and the mechanism underlying GHSR1a modulation of memory is uncertain. Here, we first report that global GHSR1a knockout enhances hippocampus-dependent memory, facilitates initial LTP in dorsal hippocampal Schaffer Collateral-CA1 synapses, and downregulates Akt activity in the hippocampus. Moreover, we show that the intrinsic excitability of GAD67+ interneurons-rather than neighboring pyramidal neurons in the dCA1-is suppressed by GHSR1a deletion, an effect that is antagonized by acute application of the Akt activator SC79. In addition, the inhibitory postsynaptic currents (IPSCs) on dCA1 pyramidal neurons are selectively reduced in mice with a GHSR1a deficiency. Finally, we demonstrate that selectively increasing the excitability of parvalbumin-expressing interneurons by hM3Dq-DREADDs increases IPSCs on dCA1 pyramidal neurons and normalizes memory in Ghsr1a KO mice. Our findings thus reveal a novel mechanism underlying memory enhancement of GHSR1a deficiency and herein support an adverse effect of GHSR1a signaling in hippocampus-dependent memory processes.
Collapse
Affiliation(s)
- Na Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Medicine, Qingdao Binhai University, 425 West Jialing River Rd, Qingdao, Shandong, 266555, China
| | - Nan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, 17 Shandong Rd, Qingdao, Shandong, 266000, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Huating Gu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Junjie Ji
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Hao Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Qianqian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
| | - Ming Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, 17 Shandong Rd, Qingdao, Shandong, 266000, China
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, 750 Agronomy Rd, College Station, TX, 77843, United States
| | - Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, 308 Ningxia Rd, Qingdao, Shandong, 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, 17 Shandong Rd, Qingdao, Shandong, 266000, China
- Department of Physiology, Institute of Brain Sciences and Related Disorders, Qingdao University, 308 Ningxia Rd., Qingdao, Shandong, 266071, China
- Department of rehabilitation medicine, Affiliated Hospital of Qingdao University, 16 Jiangsu Rd., Qingdao, Shandong, 266000, China
| |
Collapse
|
18
|
Jeczmien-Lazur JS, Sanetra AM, Pradel K, Izowit G, Chrobok L, Palus-Chramiec K, Piggins HD, Lewandowski MH. Metabolic cues impact non-oscillatory intergeniculate leaflet and ventral lateral geniculate nucleus: standard versus high-fat diet comparative study. J Physiol 2023; 601:979-1016. [PMID: 36661095 DOI: 10.1113/jp283757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) are subcortical structures involved in entrainment of the brain's circadian system to photic and non-photic (e.g. metabolic and arousal) cues. Both receive information about environmental light from photoreceptors, exhibit infra-slow oscillations (ISO) in vivo, and connect to the master circadian clock. Although current evidence demonstrates that the IGL/VLG communicate metabolic information and are crucial for entrainment of circadian rhythms to time-restricted feeding, their sensitivity to food intake-related peptides has not been investigated yet. We examined the effect of metabolically relevant peptides on the spontaneous activity of IGL/VLG neurons. Using ex vivo and in vivo electrophysiological recordings as well as in situ hybridisation, we tested potential sensitivity of the IGL/VLG to anorexigenic and orexigenic peptides, such as cholecystokinin, glucagon-like peptide 1, oxyntomodulin, peptide YY, orexin A and ghrelin. We explored neuronal responses to these drugs during day and night, and in standard vs. high-fat diet conditions. We found that IGL/VLG neurons responded to all the substances tested, except peptide YY. Moreover, more neurons responded to anorexigenic drugs at night, while a high-fat diet affected the IGL/VLG sensitivity to orexigenic peptides. Interestingly, ISO neurons responded to light and orexin A, but did not respond to the other food intake-related peptides. In contrast, non-ISO cells were activated by metabolic peptides, with only some being responsive to light. Our results show for the first time that peptides involved in the body's energy homeostasis stimulate the thalamus and suggest functional separation of the IGL/VLG cells. KEY POINTS: The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) of the rodent thalamus process various signals and participate in circadian entrainment. In both structures, cells exhibiting infra-slow oscillatory activity as well as non-rhythmically firing neurons being observed. Here, we reveal that only one of these two groups of cells responds to anorexigenic (cholecystokinin, glucagon-like peptide 1 and oxyntomodulin) and orexigenic (ghrelin and orexin A) peptides. Neuronal responses vary depending on the time of day (day vs. night) and on the diet (standard vs. high-fat diet). Additionally, we visualised receptors to the tested peptides in the IGL/VLG using in situ hybridisation. Our results suggest that two electrophysiologically different subpopulations of IGL/VLG neurons are involved in two separate functions: one related to the body's energy homeostasis and one associated with the subcortical visual system.
Collapse
Affiliation(s)
- Jagoda S Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Anna M Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Gabriela Izowit
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland.,School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Hugh D Piggins
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Marian H Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
19
|
Conde K, Kulyk D, Vanschaik A, Daisey S, Rojas C, Wiersielis K, Yasrebi A, Degroat TJ, Sun Y, Roepke TA. Deletion of Growth Hormone Secretagogue Receptor in Kisspeptin Neurons in Female Mice Blocks Diet-Induced Obesity. Biomolecules 2022; 12:1370. [PMID: 36291579 PMCID: PMC9599822 DOI: 10.3390/biom12101370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 01/19/2023] Open
Abstract
The gut peptide, ghrelin, mediates energy homeostasis and reproduction by acting through its receptor, growth hormone secretagogue receptor (GHSR), expressed in hypothalamic neurons in the arcuate (ARC). We have shown 17β-estradiol (E2) increases Ghsr expression in Kisspeptin/Neurokinin B/Dynorphin (KNDy) neurons, enhancing sensitivity to ghrelin. We hypothesized that E2-induced Ghsr expression augments KNDy sensitivity in a fasting state by elevating ghrelin to disrupt energy expenditure in females. We produced a Kiss1-GHSR knockout to determine the role of GHSR in ARC KNDy neurons. We found that changes in ARC gene expression with estradiol benzoate (EB) treatment were abrogated by the deletion of GHSR and ghrelin abolished these differences. We also observed changes in metabolism and fasting glucose levels. Additionally, knockouts were resistant to body weight gain on a high fat diet (HFD). Behaviorally, we found that knockouts on HFD exhibited reduced anxiety-like behavior. Furthermore, knockouts did not refeed to the same extent as controls after a 24 h fast. Finally, in response to cold stress, knockout females had elevated metabolic parameters compared to controls. These data indicate GHSR in Kiss1 neurons modulate ARC gene expression, metabolism, glucose homeostasis, behavior, and thermoregulation, illustrating a novel mechanism for E2 and ghrelin to control Kiss1 neurons.
Collapse
Affiliation(s)
- Kristie Conde
- Graduate Program in Neuroscience, Rutgers University Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Danielle Kulyk
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Allison Vanschaik
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Sierra Daisey
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Catherine Rojas
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Kimberly Wiersielis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Thomas J. Degroat
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Troy A. Roepke
- Graduate Program in Neuroscience, Rutgers University Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Rutgers Center for Lipid Research, the Center for Nutrition, Microbiome, and Health, and the New Jersey Institute of Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
20
|
Ma Y, Zhang H, Guo W, Yu L. Potential role of ghrelin in the regulation of inflammation. FASEB J 2022; 36:e22508. [PMID: 35983825 DOI: 10.1096/fj.202200634r] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022]
Abstract
Several diseases are caused or progress due to inflammation. In the past few years, accumulating evidence suggests that ghrelin, a gastric hormone of 28-amino acid residue length, exerts protective effects against inflammation by modulating the related pathways. This review focuses on ghrelin's anti-inflammatory and potential therapeutic effects in neurological, cardiovascular, respiratory, hepatic, gastrointestinal, and kidney disorders. Ghrelin significantly alleviates excessive inflammation and reduces damage to different target organs mainly by reducing the secretion of inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α), and inhibiting the nuclear factor kappa-B (NF-κB) and NLRP3 inflammasome signaling pathways. Ghrelin also regulates inflammation and apoptosis through the p38 MAPK/c-Jun N-terminal kinase (JNK) signaling pathway; restores cerebral microvascular integrity, and attenuates vascular leakage. Ghrelin activates the phosphoInositide-3 kinase (PI3K)/protein kinase B (Akt) pathway and inhibits inflammatory responses in cardiovascular diseases and acute kidney injury. Some studies show that ghrelin exacerbates colonic and intestinal manifestations of colitis. Interestingly, some inflammatory states, such as non-alcoholic steatohepatitis, inflammatory bowel diseases, and chronic kidney disease, are often associated with high ghrelin levels. Thus, ghrelin may be a potential new therapeutic target for inflammation-related diseases.
Collapse
Affiliation(s)
- Yunxiao Ma
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haifeng Zhang
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Weiying Guo
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lu Yu
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
21
|
Fernandez G, Cabral A, De Francesco PN, Uriarte M, Reynaldo M, Castrogiovanni D, Zubiría G, Giovambattista A, Cantel S, Denoyelle S, Fehrentz JA, Tolle V, Schiöth HB, Perello M. GHSR controls food deprivation-induced activation of CRF neurons of the hypothalamic paraventricular nucleus in a LEAP2-dependent manner. Cell Mol Life Sci 2022; 79:277. [PMID: 35504998 PMCID: PMC11072678 DOI: 10.1007/s00018-022-04302-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Prolonged fasting is a major challenge for living organisms. An appropriate metabolic response to food deprivation requires the activation of the corticotropin-releasing factor-producing neurons of the hypothalamic paraventricular nucleus (PVHCRF neurons), which are a part of the hypothalamic-pituitary-adrenal axis (HPA), as well as the growth hormone secretagogue receptor (GHSR) signaling, whose activity is up- or down-regulated, respectively, by the hormones ghrelin and the liver-expressed antimicrobial peptide 2 (LEAP2). Since ghrelin treatment potently up-regulates the HPA axis, we studied the role of GHSR in mediating food deprivation-induced activation of the PVHCRF neurons in mice. METHODS We estimated the activation of the PVHCRF neurons, using immuno-staining against CRF and the marker of neuronal activation c-Fos in brain sections, and assessed plasma levels of corticosterone and glucose in different pharmacologically or genetically manipulated mouse models exposed, or not, to a 2-day food deprivation protocol. In particular, we investigated ad libitum fed or food-deprived male mice that: (1) lacked GHSR gene expression, (2) had genetic deletion of the ghrelin gene, (3) displayed neurotoxic ablation of the hypothalamic arcuate nucleus, (4) were centrally treated with an anti-ghrelin antibody to block central ghrelin action, (5) were centrally treated with a GHSR ligand that blocks ghrelin-evoked and constitutive GHSR activities, or (6) received a continuous systemic infusion of LEAP2(1-12). RESULTS We found that food deprivation results in the activation of the PVHCRF neurons and in a rise of the ghrelin/LEAP2 molar ratio. Food deprivation-induced activation of PVHCRF neurons required the presence and the signaling of GHSR at hypothalamic level, but not of ghrelin. Finally, we found that preventing the food deprivation-induced fall of LEAP2 reverses the activation of the PVHCRF neurons in food-deprived mice, although it has no effect on body weight or blood glucose. CONCLUSION Food deprivation-induced activation of the PVHCRF neurons involves ghrelin-independent actions of GHSR at hypothalamic level and requires a decrease of plasma LEAP2 levels. We propose that the up-regulation of the actions of GHSR associated to the fall of plasma LEAP2 level are physiologically relevant neuroendocrine signals during a prolonged fasting.
Collapse
Affiliation(s)
- Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Agustina Cabral
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Maia Uriarte
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Mirta Reynaldo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Daniel Castrogiovanni
- Cell Culture Facility, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Guillermina Zubiría
- Laboratory of Neuroendocrinology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Andrés Giovambattista
- Laboratory of Neuroendocrinology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina
| | - Sonia Cantel
- Institut Des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, France
| | - Severine Denoyelle
- Institut Des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut Des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM, Montpellier, France
| | - Virginie Tolle
- Institute of Psychiatry and Neuroscience of Paris, Université de Paris, UMR-S 1266 INSERM, Paris, France
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
- Institute for Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata (UNLP)], Calle 526 S/N entre 10 y 11, La Plata, Buenos Aires, 1900, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
22
|
Islam MN, Zhang W, Sakai K, Nakazato Y, Tanida R, Sakoda H, Takei T, Takao T, Nakazato M. Liver-expressed antimicrobial peptide 2 functions independently of growth hormone secretagogue receptor in calorie-restricted mice. Peptides 2022; 151:170763. [PMID: 35151767 DOI: 10.1016/j.peptides.2022.170763] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022]
Abstract
Ghrelin is a gastric-derived peptide that stimulates feeding, blood glucose elevation, body temperature reduction, and growth hormone (GH) secretion. Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous antagonist of the ghrelin receptor, also called growth hormone secretagogue receptor (GHSR). We studied the effects of LEAP2 administration on feeding, body weight, glycemia, body temperature, and inflammation-related genes in the liver in C57BL/6 J mice and Ghsr-knockout (Ghsr-KO) mice. We found that a single administration of LEAP2 did not abolish fasting-induced food intake in 24-h fasted C57BL/6 J mice or Ghsr-KO mice. Moreover, continuous LEAP2 administration to mice fed ad libitum for 6 days did not affect feeding, body temperature, plasma ghrelin, or blood glucose. By contrast, continuous LEAP2 administration to calorie-restricted C57BL/6 J mice and Ghsr-KO mice induced body weight loss, hypoglycemia, body temperature reduction, and upregulation of Il-6 and Il-1β mRNAs in the liver. Our findings suggest that LEAP2 functions independently of GHSR, implying that LEAP2 affects physiology beyond the ghrelin-GHSR system.
Collapse
Affiliation(s)
- Md Nurul Islam
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Weidong Zhang
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Katsuya Sakai
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuki Nakazato
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Ryota Tanida
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Ishikawa 920-8640, Japan
| | - Hideyuki Sakoda
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Toshiki Takei
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Toshifumi Takao
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Masamitsu Nakazato
- Division of Interactive Organ Systems, Department of Projects Research, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan; AMED-CREST, Agency for Medical Research and Development, Tokyo 100-0004, Japan.
| |
Collapse
|
23
|
Mechanistic Investigation of GHS-R Mediated Glucose-Stimulated Insulin Secretion in Pancreatic Islets. Biomolecules 2022; 12:biom12030407. [PMID: 35327599 PMCID: PMC8945998 DOI: 10.3390/biom12030407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/19/2022] [Accepted: 02/27/2022] [Indexed: 02/07/2023] Open
Abstract
Ghrelin receptor, a growth hormone secretagogue receptor (GHS-R), is expressed in the pancreas. Emerging evidence indicates that GHS-R is involved in the regulation of glucose-stimulated insulin secretion (GSIS), but the mechanism by which GHS-R regulates GSIS in the pancreas is unclear. In this study, we investigated the role of GHS-R on GSIS in detail using global Ghsr−/− mice (in vivo) and Ghsr-ablated pancreatic islets (ex vivo). GSIS was attenuated in both Ghsr−/− mice and Ghsr-ablated islets, while the islet morphology was similar between WT and Ghsr−/− mice. To elucidate the mechanism underpinning Ghsr-mediated GSIS, we investigated the key steps of the GSIS signaling cascade. The gene expression of glucose transporter 2 (Glut2) and the glucose-metabolic intermediate—glucose-6-phosphate (G6P) were reduced in Ghsr-ablated islets, supporting decreased glucose uptake. There was no difference in mitochondrial DNA content in the islets of WT and Ghsr−/− mice, but the ATP/ADP ratio in Ghsr−/− islets was significantly lower than that of WT islets. Moreover, the expression of pancreatic and duodenal homeobox 1 (Pdx1), as well as insulin signaling genes of insulin receptor (IR) and insulin receptor substrates 1 and 2 (IRS1/IRS2), was downregulated in Ghsr−/− islets. Akt is the key mediator of the insulin signaling cascade. Concurrently, Akt phosphorylation was reduced in the pancreas of Ghsr−/− mice under both insulin-stimulated and homeostatic conditions. These findings demonstrate that GHS-R ablation affects key components of the insulin signaling pathway in the pancreas, suggesting the existence of a cross-talk between GHS-R and the insulin signaling pathway in pancreatic islets, and GHS-R likely regulates GSIS via the Akt-Pdx1-GLUT2 pathway.
Collapse
|
24
|
Chen X, Dong J, Jiao Q, Du X, Bi M, Jiang H. "Sibling" battle or harmony: crosstalk between nesfatin-1 and ghrelin. Cell Mol Life Sci 2022; 79:169. [PMID: 35239020 PMCID: PMC11072372 DOI: 10.1007/s00018-022-04193-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/17/2022]
Abstract
Ghrelin was first identified as an endogenous ligand of the growth hormone secretagogue receptor (GHSR) in 1999, with the function of stimulating the release of growth hormone (GH), while nesfatin-1 was identified in 2006. Both peptides are secreted by the same kind of endocrine cells, X/A-like cells in the stomach. Compared with ghrelin, nesfatin-1 exerts opposite effects on energy metabolism, glucose metabolism, gastrointestinal functions and regulation of blood pressure, but exerts similar effects on anti-inflammation and neuroprotection. Up to now, nesfatin-1 remains as an orphan ligand because its receptor has not been identified. Several studies have shown the effects of nesfatin-1 are dependent on the receptor of ghrelin. We herein compare the effects of nesfatin-1 and ghrelin in several aspects and explore the possibility of their interactions.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Jing Dong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
25
|
Müller TD, Blüher M, Tschöp MH, DiMarchi RD. Anti-obesity drug discovery: advances and challenges. Nat Rev Drug Discov 2022; 21:201-223. [PMID: 34815532 PMCID: PMC8609996 DOI: 10.1038/s41573-021-00337-8] [Citation(s) in RCA: 525] [Impact Index Per Article: 175.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2021] [Indexed: 12/27/2022]
Abstract
Enormous progress has been made in the last half-century in the management of diseases closely integrated with excess body weight, such as hypertension, adult-onset diabetes and elevated cholesterol. However, the treatment of obesity itself has proven largely resistant to therapy, with anti-obesity medications (AOMs) often delivering insufficient efficacy and dubious safety. Here, we provide an overview of the history of AOM development, focusing on lessons learned and ongoing obstacles. Recent advances, including increased understanding of the molecular gut-brain communication, are inspiring the pursuit of next-generation AOMs that appear capable of safely achieving sizeable and sustained body weight loss.
Collapse
Affiliation(s)
- Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | | |
Collapse
|
26
|
Lugilde J, Casado S, Beiroa D, Cuñarro J, Garcia-Lavandeira M, Álvarez CV, Nogueiras R, Diéguez C, Tovar S. LEAP-2 Counteracts Ghrelin-Induced Food Intake in a Nutrient, Growth Hormone and Age Independent Manner. Cells 2022; 11:cells11030324. [PMID: 35159134 PMCID: PMC8834077 DOI: 10.3390/cells11030324] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/30/2022] Open
Abstract
Data gleaned recently shows that ghrelin, a stomach derived peptide, and liver-expressed-antimicrobial peptide 2 (LEAP-2) play opposite roles on food intake. However, the data available with LEAP-2 in relation to in vivo studies are still very scanty and some key questions regarding the interplay among ghrelin and LEAP-2 remain to be answered. In this work, using rats and mice, we study fasting-induced food intake as well as testing the effect of diet exposure, e.g., standard diet and high fat diet, in terms of ghrelin-induced food intake. The anorexigenic effect of LEAP-2 on fasting induced food intake appears to be dependent on energy stores, being more evident in ob/ob than in wild type mice and also in animals exposed to high fat diet. On the other hand, LEAP-2 administration markedly inhibited ghrelin-induced food intake in lean, obese (ob/ob and DIO) mice, aged rats and GH-deficient dwarf rats. In contrast, the inhibitory effect on glucose levels can only be observed in some specific experimental models indicating that the mechanisms involved are likely to be quite different. Taken together from these data, LEAP-2 emerged as a potential candidate to be therapeutically useful in obesity.
Collapse
Affiliation(s)
- Javier Lugilde
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
| | - Sabela Casado
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Daniel Beiroa
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
| | - Juan Cuñarro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
| | - Montserrat Garcia-Lavandeira
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain; (M.G.-L.); (C.V.Á.)
| | - Clara V. Álvarez
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), 15782 Santiago de Compostela, Spain; (M.G.-L.); (C.V.Á.)
| | - Rubén Nogueiras
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Carlos Diéguez
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (C.D.); (S.T.)
| | - Sulay Tovar
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (J.L.); (S.C.); (D.B.); (J.C.); (R.N.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (C.D.); (S.T.)
| |
Collapse
|
27
|
Vohra MS, Benchoula K, Serpell CJ, Hwa WE. AgRP/NPY and POMC neurons in the arcuate nucleus and their potential role in treatment of obesity. Eur J Pharmacol 2022; 915:174611. [PMID: 34798121 DOI: 10.1016/j.ejphar.2021.174611] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity is a major health crisis affecting over a third of the global population. This multifactorial disease is regulated via interoceptive neural circuits in the brain, whose alteration results in excessive body weight. Certain central neuronal populations in the brain are recognised as crucial nodes in energy homeostasis; in particular, the hypothalamic arcuate nucleus (ARC) region contains two peptide microcircuits that control energy balance with antagonistic functions: agouti-related peptide/neuropeptide-Y (AgRP/NPY) signals hunger and stimulates food intake; and pro-opiomelanocortin (POMC) signals satiety and reduces food intake. These neuronal peptides levels react to energy status and integrate signals from peripheral ghrelin, leptin, and insulin to regulate feeding and energy expenditure. To manage obesity comprehensively, it is crucial to understand cellular and molecular mechanisms of information processing in ARC neurons, since these regulate energy homeostasis. Importantly, a specific strategy focusing on ARC circuits needs to be devised to assist in treating obese patients and maintaining weight loss with minimal or no side effects. The aim of this review is to elucidate the recent developments in the study of AgRP-, NPY- and POMC-producing neurons, specific to their role in controlling metabolism. The impact of ghrelin, leptin, and insulin signalling via action of these neurons is also surveyed, since they also impact energy balance through this route. Lastly, we present key proteins, targeted genes, compounds, drugs, and therapies that actively work via these neurons and could potentially be used as therapeutic targets for treating obesity conditions.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Wong Eng Hwa
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
28
|
Molecular mechanism of agonism and inverse agonism in ghrelin receptor. Nat Commun 2022; 13:300. [PMID: 35027551 PMCID: PMC8758724 DOI: 10.1038/s41467-022-27975-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
Much effort has been invested in the investigation of the structural basis of G protein-coupled receptors (GPCRs) activation. Inverse agonists, which can inhibit GPCRs with constitutive activity, are considered useful therapeutic agents, but the molecular mechanism of such ligands remains insufficiently understood. Here, we report a crystal structure of the ghrelin receptor bound to the inverse agonist PF-05190457 and a cryo-electron microscopy structure of the active ghrelin receptor-Go complex bound to the endogenous agonist ghrelin. Our structures reveal a distinct binding mode of the inverse agonist PF-05190457 in the ghrelin receptor, different from the binding mode of agonists and neutral antagonists. Combining the structural comparisons and cellular function assays, we find that a polar network and a notable hydrophobic cluster are required for receptor activation and constitutive activity. Together, our study provides insights into the detailed mechanism of ghrelin receptor binding to agonists and inverse agonists, and paves the way to design specific ligands targeting ghrelin receptors. Ghrelin receptor regulates energy homeostasis through constitutive activity or by the ghrelin. Here the authors report two structures of ghrelin receptor bound to agonist and inverse agonist, providing insights into the mechanism of inverse agonism, which is of interest for specific ligand design.
Collapse
|
29
|
Petersen N, Greiner TU, Torz L, Bookout A, Gerstenberg MK, Castorena CM, Kuhre RE. Targeting the Gut in Obesity: Signals from the Inner Surface. Metabolites 2022; 12:metabo12010039. [PMID: 35050161 PMCID: PMC8778595 DOI: 10.3390/metabo12010039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/26/2021] [Accepted: 12/31/2021] [Indexed: 12/17/2022] Open
Abstract
Obesity is caused by prolonged energy surplus. Current anti-obesity medications are mostly centralized around the energy input part of the energy balance equation by increasing satiety and reducing appetite. Our gastrointestinal tract is a key organ for regulation of food intake and supplies a tremendous number of circulating signals that modulate the activity of appetite-regulating areas of the brain by either direct interaction or through the vagus nerve. Intestinally derived messengers are manifold and include absorbed nutrients, microbial metabolites, gut hormones and other enterokines, collectively comprising a fine-tuned signalling system to the brain. After a meal, nutrients directly interact with appetite-inhibiting areas of the brain and induce satiety. However, overall feeding behaviour also depends on secretion of gut hormones produced by highly specialized and sensitive enteroendocrine cells. Moreover, circulating microbial metabolites and their interactions with enteroendocrine cells further contribute to the regulation of feeding patterns. Current therapies exploiting the appetite-regulating properties of the gut are based on chemically modified versions of the gut hormone, glucagon-like peptide-1 (GLP-1) or on inhibitors of the primary GLP-1 inactivating enzyme, dipeptidyl peptidase-4 (DPP-4). The effectiveness of these approaches shows that that the gut is a promising target for therapeutic interventions to achieve significant weigh loss. We believe that increasing understanding of the functionality of the intestinal epithelium and new delivery systems will help develop selective and safe gut-based therapeutic strategies for improved obesity treatment in the future. Here, we provide an overview of the major homeostatic appetite-regulating signals generated by the intestinal epithelial cells and how these signals may be harnessed to treat obesity by pharmacological means.
Collapse
Affiliation(s)
- Natalia Petersen
- Global Obesity and Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Novo Park 1, 2670 Måløv, Denmark; (L.T.); (M.K.G.); (R.E.K.)
- Correspondence:
| | - Thomas U. Greiner
- The Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden;
| | - Lola Torz
- Global Obesity and Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Novo Park 1, 2670 Måløv, Denmark; (L.T.); (M.K.G.); (R.E.K.)
- Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Angie Bookout
- Global Obesity and Liver Disease Research, Global Drug Discovery, Novo Nordisk Research Center, Seattle, WA 98109, USA; (A.B.); (C.M.C.)
| | - Marina Kjærgaard Gerstenberg
- Global Obesity and Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Novo Park 1, 2670 Måløv, Denmark; (L.T.); (M.K.G.); (R.E.K.)
| | - Carlos M. Castorena
- Global Obesity and Liver Disease Research, Global Drug Discovery, Novo Nordisk Research Center, Seattle, WA 98109, USA; (A.B.); (C.M.C.)
| | - Rune Ehrenreich Kuhre
- Global Obesity and Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Novo Park 1, 2670 Måløv, Denmark; (L.T.); (M.K.G.); (R.E.K.)
- Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
30
|
Labarthe A, Zizzari P, Fiquet O, Lebrun N, Veldhuis JD, Roelfsema F, Chauveau C, Bohlooly-Y M, Epelbaum J, Tolle V. Effect of Growth Hormone Secretagogue Receptor Deletion on Growth, Pulsatile Growth Hormone Secretion, and Meal Pattern in Male and Female Mice. Neuroendocrinology 2022; 112:215-234. [PMID: 33774644 DOI: 10.1159/000516147] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/25/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION While the vast majority of research investigating the role of ghrelin or its receptor, GHS-R1a, in growth, feeding, and metabolism has been conducted in male rodents, very little is known about sex differences in this system. Furthermore, the role of GHS-R1a signaling in the control of pulsatile GH secretion and its link with growth or metabolic parameters has never been characterized. METHODS We assessed the sex-specific contribution of GHS-R1a signaling in the activity of the GH/IGF-1 axis, metabolic parameters, and feeding behavior in adolescent (5-6 weeks old) or adult (10-19 weeks old) GHS-R KO (Ghsr-/-) and WT (Ghsr+/+) male and female mice. RESULTS Adult Ghsr-/- male and female mice displayed deficits in weight and linear growth that were correlated with reduced GH pituitary contents in males only. GHS-R1a deletion was associated with reduced meal frequency and increased meal intervals, as well as reduced hypothalamic GHRH and NPY mRNA in males, not females. In adult, GH release from Ghsr-/- mice pituitary explants ex vivo was reduced independently of the sex. However, in vivo pulsatile GH secretion decreased in adult but not adolescent Ghsr-/- females, while in males, GHS-R1a deletion was associated with reduction in pulsatile GH secretion during adolescence exclusively. In males, linear growth did not correlate with pulsatile GH secretion, but rather with ApEn, a measure that reflects irregularity of the rhythmic secretion. Fat mass, plasma leptin concentrations, or ambulatory activity did not predict differences in GH secretion. DISCUSSION/CONCLUSION These results point to a sex-dependent dimorphic effect of GHS-R1a signaling to modulate pulsatile GH secretion and meal pattern in mice with different compensatory mechanisms occurring in the hypothalamus of adult males and females after GHS-R1a deletion. Altogether, we show that GHS-R1a signaling plays a more critical role in the regulation of pulsatile GH secretion during adolescence in males and adulthood in females.
Collapse
Affiliation(s)
- Alexandra Labarthe
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Philippe Zizzari
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Oriane Fiquet
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Nicolas Lebrun
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Johannes D Veldhuis
- Endocrine Research Unit, Department of Medicine, Mayo School of Graduate Medical Education, Clinical Translational Science Center, Mayo Clinic, Rochester, New York, USA
| | - Ferdinand Roelfsema
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, The Netherlands
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab - MABLab ULR 4490, University Littoral Côte d'Opale, Boulogne-sur-Mer, France
- University Lille, CHU Lille, Lille, France
| | - Mohammad Bohlooly-Y
- Translational Genomics, Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jacques Epelbaum
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, Brunoy, France
| | - Virginie Tolle
- Université de Paris, UMRS_1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| |
Collapse
|
31
|
Peris-Sampedro F, Le May MV, Stoltenborg I, Schéle E, Dickson SL. A skeleton in the cupboard in ghrelin research: Where are the skinny dwarfs? J Neuroendocrinol 2021; 33:e13025. [PMID: 34427011 DOI: 10.1111/jne.13025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/09/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022]
Abstract
Based on studies delivering ghrelin or ghrelin receptor agonists, we have learned a great deal about the importance of the brain ghrelin signalling system for a wide range of physiological processes that include feeding behaviours, growth hormone secretion and glucose homeostasis. Because these processes can be considered as essential to life, the question arises as to why mouse models of depleted ghrelin signalling are not all skinny dwarfs with a host of behavioural and metabolic problems. Here, we provide a systematic detailed review of the phenotype of mice with deficient ghrelin signalling to help better understand the relevance and importance of the brain ghrelin signalling system, with a particular emphasis on those questions that remain unanswered.
Collapse
Affiliation(s)
- Fiona Peris-Sampedro
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie V Le May
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Iris Stoltenborg
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Schéle
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Suzanne L Dickson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
32
|
Hassouna R, Fernandez G, Lebrun N, Fiquet O, Roelfsema F, Labarthe A, Zizzari P, Tomasetto C, Epelbaum J, Viltart O, Chauveau C, Perello M, Tolle V. Ghrelin Gene Deletion Alters Pulsatile Growth Hormone Secretion in Adult Female Mice. Front Endocrinol (Lausanne) 2021; 12:754522. [PMID: 34721302 PMCID: PMC8549963 DOI: 10.3389/fendo.2021.754522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Using preproghrelin-deficient mice (Ghrl-/-), we previously observed that preproghrelin modulates pulsatile growth hormone (GH) secretion in post-pubertal male mice. However, the role of ghrelin and its derived peptides in the regulation of growth parameters or feeding in females is unknown. We measured pulsatile GH secretion, growth, metabolic parameters and feeding behavior in adult Ghrl-/- and Ghrl+/+ male and female mice. We also assessed GH release from pituitary explants and hypothalamic growth hormone-releasing hormone (GHRH) expression and immunoreactivity. Body weight and body fat mass, linear growth, spontaneous food intake and food intake following a 48-h fast, GH pituitary contents and GH release from pituitary explants ex vivo, fasting glucose and glucose tolerance were not different among adult Ghrl-/- and Ghrl+/+ male or female mice. In vivo, pulsatile GH secretion was decreased, while approximate entropy, that quantified orderliness of secretion, was increased in adult Ghrl-/- females only, defining more irregular GH pattern. The number of neurons immunoreactive for GHRH visualized in the hypothalamic arcuate nucleus was increased in adult Ghrl-/- females, as compared to Ghrl+/+ females, whereas the expression of GHRH was not different amongst groups. Thus, these results point to sex-specific effects of preproghrelin gene deletion on pulsatile GH secretion, but not feeding, growth or metabolic parameters, in adult mice.
Collapse
Affiliation(s)
- Rim Hassouna
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Nicolas Lebrun
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Oriane Fiquet
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Ferdinand Roelfsema
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, Netherlands
| | - Alexandra Labarthe
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Philippe Zizzari
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Catherine Tomasetto
- Institut de génétique et de biologie moléculaire et cellulaire (IGBMC), UMR7104 CNRS/U1258 INSERM, Université de Strasbourg, Illkirch, France
| | - Jacques Epelbaum
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, Brunoy, France
| | - Odile Viltart
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
- Université de Lille, Faculté des Sciences et Technologies, Villeneuve d’Ascq, France
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab - MABLab ULR 4490, Univ. Littoral Côte d’Opale, Boulogne-sur-Mer, Univ. Lille and CHU Lille, Lille, France
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA). National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Virginie Tolle
- Université de Paris, UMR-S 1266 INSERM, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| |
Collapse
|
33
|
Liu H, Zang P, Lee I(I, Anderson B, Christiani A, Strait‐Bodey L, Breckheimer BA, Storie M, Tewnion A, Krumm K, Li T, Irwin B, Garcia JM. Growth hormone secretagogue receptor-1a mediates ghrelin's effects on attenuating tumour-induced loss of muscle strength but not muscle mass. J Cachexia Sarcopenia Muscle 2021; 12:1280-1295. [PMID: 34264027 PMCID: PMC8517358 DOI: 10.1002/jcsm.12743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/11/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ghrelin may ameliorate cancer cachexia (CC) by preventing anorexia, muscle, and fat loss. However, the mechanisms mediating these effects are not fully understood. This study characterizes the pathways involved in muscle mass and strength loss in the Lewis lung carcinoma (LLC)-induced cachexia model, and the effects of ghrelin in mice with or without its only known receptor: the growth hormone secretagogue receptor-1a ((GHSR-1a), Ghsr+/+ and Ghsr-/- ). METHODS Five to 7-month-old male C57BL/6J Ghsr+/+ and Ghsr-/- mice were inoculated with 1 × 106 heat-killed (HK) or live LLC cells (tumour implantation, TI). When tumours were palpable (7 days after TI), tumour-bearing mice were injected with vehicle (T + V) or ghrelin twice/day for 14 days (T + G, 0.8 mg/kg), while HK-treated mice were given vehicle (HK + V). Body weight and grip strength were evaluated before TI and at termination (21 days after TI). Hindlimb muscles were collected for analysis. RESULTS Less pronounced body weight (BW) loss (87.70 ± 0.98% vs. 83.92 ± 1.23%, percentage of baseline BW in tumour-bearing Ghsr+/+ vs. Ghsr-/- , P = 0.008), and lower upregulation of ubiquitin-proteasome system (UPS, MuRF1/Trim63, 5.71 ± 1.53-fold vs. 9.22 ± 1.94-fold-change from Ghsr+/+ HK + V in tumour-bearing Ghsr+/+ vs. Ghsr-/- , P = 0.036) and autophagy markers (Becn1, Atg5, Atg7, tumour-bearing Ghsr+/+ < Ghsr-/- , all P < 0.02) were found in T + V Ghsr+/+ vs. Ghsr-/- mice. Ghrelin attenuated LLC-induced UPS marker upregulation in both genotypes, [Trim63 was decreased from 5.71 ± 1.53-fold to 1.96 ± 0.47-fold in Ghsr+/+ (T + V vs. T + G: P = 0.032) and 9.22 ± 1.94-fold to 4.72 ± 1.06-fold in Ghsr-/- (T + V vs. T + G: P = 0.008)]. Only in Ghsr+/+ mice ghrelin ameliorated LLC-induced grip strength loss [improved from 89.24 ± 3.48% to 97.80 ± 2.31% of baseline (T + V vs. T + G: P = 0.042)], mitophagy markers [Bnip3 was decreased from 2.28 ± 0.56 to 1.38 ± 0.14-fold (T + V vs. T + G: P ≤ 0.05)], and impaired mitochondrial respiration [State 3u improved from 698.23 ± 73.96 to 934.37 ± 95.21 pmol/min (T + V vs. T + G: P ≤ 0.05)], whereas these markers were not improved by ghrelin Ghsr-/- . Compared with Ghsr+/+ , Ghsr-/- tumour-bearing mice also showed decreased response to ghrelin in BW [T + G-treated Ghsr+/+ vs. Ghsr -/- : 91.75 ± 1.05% vs. 86.18 ± 1.13% of baseline BW, P < 0.001)], gastrocnemius (T + G-treated Ghsr+/+ vs. Ghsr-/- : 96.9 ± 2.08% vs. 88.15 ± 1.78% of Ghsr+/+ HK + V, P < 0.001) and quadriceps muscle mass (T + G-treated Ghsr+/+ vs. Ghsr-/- : 96.12 ± 2.31% vs. 88.36 ± 1.94% of Ghsr+/+ HK + V, P = 0.01), and gastrocnemius type IIA (T + G-treated Ghsr+/+ vs. Ghsr-/- : 1250.49 ± 31.72 vs. 1017.62 ± 70.99 μm2 , P = 0.027) and IIB fibre cross-sectional area (T + G-treated Ghsr+/+ vs. Ghsr-/- : 2496.48 ± 116.88 vs. 2183.04 ± 103.43 μm2 , P = 0.024). CONCLUSIONS Growth hormone secretagogue receptor-1a mediates ghrelin's effects on attenuating LLC-induced weakness but not muscle mass loss by modulating the autophagy-lysosome pathway, mitophagy, and mitochondrial respiration.
Collapse
Affiliation(s)
- Haiming Liu
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Pu Zang
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
- Department of EndocrinologyNanjing Jinling HospitalNanjingChina
| | - Ian (In‐gi) Lee
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Barbara Anderson
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Anthony Christiani
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Lena Strait‐Bodey
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Beatrice A. Breckheimer
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Mackenzie Storie
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Alison Tewnion
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Kora Krumm
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Theresa Li
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Brynn Irwin
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| | - Jose M. Garcia
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWAUSA
- Gerontology and Geriatric MedicineUniversity of Washington Department of MedicineSeattleWAUSA
| |
Collapse
|
34
|
Peris-Sampedro F, Stoltenborg I, Le May MV, Zigman JM, Adan RAH, Dickson SL. Genetic deletion of the ghrelin receptor (GHSR) impairs growth and blunts endocrine response to fasting in Ghsr-IRES-Cre mice. Mol Metab 2021; 51:101223. [PMID: 33798772 PMCID: PMC8102639 DOI: 10.1016/j.molmet.2021.101223] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The orexigenic hormone ghrelin exerts its physiological effects by binding to and activating the growth hormone secretagogue receptor (GHSR). The recent development of a Ghsr-IRES-Cre knock-in mouse line has enabled to genetically access GHSR-expressing neurons. Inserting a Cre construct using a knock-in strategy, even when following an upstream internal ribosome entry site (IRES) can, however, interfere with expression of a targeted gene, with consequences for the phenotype emerging. This study aimed to phenotype, both physically and metabolically, heterozygous and homozygous Ghsr-IRES-Cre mice, with a view to discovering the extent to which the ghrelin signalling system remains functional in these mice. METHODS We assessed feeding and arcuate nucleus (Arc) Fos activation in wild-type, heterozygous and homozygous Ghsr-IRES-Cre mice in response to peripherally-administered ghrelin. We also characterised their developmental and growth phenotypes, as well as their metabolic responses upon an overnight fast. RESULTS Insertion of the IRES-Cre cassette into the 3'-untranslated region of the Ghsr gene led to a gene-dosage GHSR depletion in the Arc. Whereas heterozygotes remained ghrelin-responsive and more closely resembled wild-types, ghrelin had reduced orexigenic efficacy and failed to induce Arc Fos expression in homozygous littermates. Homozygotes had a lower body weight accompanied by a shorter body length, less fat tissue content, altered bone parameters, and lower insulin-like growth factor-1 levels compared to wild-type and heterozygous littermates. Moreover, both heterozygous and homozygous Ghsr-IRES-Cre mice lacked the usual fasting-induced rise in growth hormone (GH) and displayed an exaggerated drop in blood glucose and insulin compared to wild-types. Unexpectedly, fasting acyl-ghrelin levels were allele-dependently increased. CONCLUSIONS Our data suggest that (i) heterozygous but not homozygous Ghsr-IRES-Cre mice retain the usual responsiveness to administered ghrelin, (ii) the impact of fasting on GH release and glucose homeostasis is altered even when only one copy of the Ghsr gene is non-functional (as in heterozygous Ghsr-IRES-Cre mice) and (iii) homozygous Ghsr-IRES-Cre mice exhibit growth retardation. Of the many transgenic models of suppressed ghrelin signalling, Ghsr-IRES-Cre mice emerge as best representing the full breadth of the expected phenotype with respect to body weight, growth, and metabolic parameters.
Collapse
Affiliation(s)
- Fiona Peris-Sampedro
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Iris Stoltenborg
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie V Le May
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Roger A H Adan
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Suzanne L Dickson
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
35
|
Nogueiras R. MECHANISMS IN ENDOCRINOLOGY: The gut-brain axis: regulating energy balance independent of food intake. Eur J Endocrinol 2021; 185:R75-R91. [PMID: 34260412 PMCID: PMC8345901 DOI: 10.1530/eje-21-0277] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Obesity is a global pandemic with a large health and economic burden worldwide. Bodyweight is regulated by the ability of the CNS, and especially the hypothalamus, to orchestrate the function of peripheral organs that play a key role in metabolism. Gut hormones play a fundamental role in the regulation of energy balance, as they modulate not only feeding behavior but also energy expenditure and nutrient partitioning. This review examines the recent discoveries about hormones produced in the stomach and gut, which have been reported to regulate food intake and energy expenditure in preclinical models. Some of these hormones act on the hypothalamus to modulate thermogenesis and adiposity in a food intake-independent fashion. Finally, the association of these gut hormones to eating, energy expenditure, and weight loss after bariatric surgery in humans is discussed.
Collapse
Affiliation(s)
- Ruben Nogueiras
- Department of Physiology, CIMUS, USC, CIBER Fisiopatología Obesidad y Nutrición (CiberOBN), Instituto Salud Carlos III, Galician Agency of Innovation, Xunta de Galicia, Santiago de Compostela, Spain
| |
Collapse
|
36
|
Leptin treatment prevents impaired hypoglycemic counterregulation induced by exposure to severe caloric restriction or exposure to recurrent hypoglycemia. Auton Neurosci 2021; 235:102853. [PMID: 34358845 DOI: 10.1016/j.autneu.2021.102853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 06/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Hypoglycemia-associated autonomic failure (HAAF) is a maladaptive failure in glucose counterregulation in persons with diabetes (PWD) that is caused by recurrent exposure to hypoglycemia. The adipokine leptin is known to regulate glucose homeostasis, and leptin levels fall following exposure to recurrent hypoglycemia. Yet, little is known regarding how reduced leptin levels influence glucose counterregulation, or if low leptin levels are involved in the development of HAAF. The purpose of this study was to determine the effect of hypoleptinemia on the neuroendocrine responses to hypoglycemia. We utilized two separate experimental paradigms known to induce a hypoleptinemic state: 60% caloric restriction (CR) in mice and three days of recurrent hypoglycemia (3dRH) in rats. A sub-set of animals were also treated with leptin (0.5-1.0 μg/g) during the CR or 3dRH periods. Neuroendocrine responses to hypoglycemia were assessed 60 min following an IP insulin injection on the terminal day of the paradigms. CR mice displayed defects in hypoglycemic counterregulation, indicated by significantly lower glucagon levels relative to controls, 13.5 pmol/L (SD 10.7) versus 64.7 pmol/L (SD 45) (p = 0.002). 3dRH rats displayed reduced epinephrine levels relative to controls, 1900 pg/mL (SD 1052) versus 3670 pg/mL (SD 780) (p = 0.030). Remarkably, leptin treatment during either paradigm completely reversed this effect by normalizing glucagon levels in CR mice, 78.0 pmol/L (SD 47.3) (p = 0.764), and epinephrine levels in 3dRH rats, 2910 pg/mL (SD 1680) (p = 0.522). These findings suggest that hypoleptinemia may be a key signaling event driving the development of HAAF and that leptin treatment may prevent the development of HAAF in PWD.
Collapse
|
37
|
|
38
|
β Cell GHS-R Regulates Insulin Secretion and Sensitivity. Int J Mol Sci 2021; 22:ijms22083950. [PMID: 33920473 PMCID: PMC8069226 DOI: 10.3390/ijms22083950] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022] Open
Abstract
Growth hormone secretagogue receptor (GHS-R) is widely known to regulate food intake and adiposity, but its role in glucose homeostasis is unclear. In this study, we investigated the expression of GHS-R in mouse pancreatic islets and its role in glycemic regulation. We used Ghsr-IRES-tauGFP mice, with Green Fluorescent Protein (GFP) as a surrogate for GHS-R, to demonstrate the GFP co-localization with insulin and glucagon expression in pancreatic islets, confirming GHS-R expression in β and α cells. We then generated β-cell-specific GHSR-deleted mice with MIP-Cre/ERT and validated that GHS-R suppression was restricted to the pancreatic islets. MIP-Cre/ERT;Ghsrf/f mice showed normal energy homeostasis with similar body weight, body composition, and indirect calorimetry profile. Interestingly, MIP-Cre/ERT;Ghsrf/f mice exhibited an impressive phenotype in glucose homeostasis. Compared to controls, MIP-Cre/ERT;Ghsrf/f mice showed lower fasting blood glucose and insulin; reduced first-phase insulin secretion during a glucose tolerance test (GTT) and glucose-stimulated insulin secretion (GSIS) test in vivo. The isolated pancreatic islets of MIP-Cre/ERT;Ghsrf/f mice also showed reduced insulin secretion during GSIS ex vivo. Further, MIP-Cre/ERT;Ghsrf/f mice exhibited improved insulin sensitivity during insulin tolerance tests (ITT). Overall, our results confirmed GHS-R expression in pancreatic β and α cells; GHS-R cell-autonomously regulated GSIS and modulated systemic insulin sensitivity. In conclusion, β cell GHS-R was an important regulator of glucose homeostasis, and GHS-R antagonists may have therapeutic potential for Type 2 Diabetes.
Collapse
|
39
|
Calder AN, Yu T, Dahir NS, Sun Y, Gilbertson TA. Ghrelin Receptors Enhance Fat Taste Responsiveness in Female Mice. Nutrients 2021; 13:nu13041045. [PMID: 33804920 PMCID: PMC8063820 DOI: 10.3390/nu13041045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023] Open
Abstract
Ghrelin is a major appetite-stimulating neuropeptide found in circulation. While its role in increasing food intake is well known, its role in affecting taste perception, if any, remains unclear. In this study, we investigated the role of the growth hormone secretagogue receptor's (GHS-R; a ghrelin receptor) activity in the peripheral taste system using feeding studies and conditioned taste aversion assays by comparing wild-type and GHS-R-knockout models. Using transgenic mice expressing enhanced green fluorescent protein (GFP), we demonstrated GHS-R expression in the taste system in relation phospholipase C ß2 isotype (PLCβ2; type II taste cell marker)- and glutamate decarboxylase type 67 (GAD67; type III taste cell marker)-expressing cells using immunohistochemistry. We observed high levels of co-localization between PLCβ2 and GHS-R within the taste system, while GHS-R rarely co-localized in GAD67-expressing cells. Additionally, following 6 weeks of 60% high-fat diet, female Ghsr-/- mice exhibited reduced responsiveness to linoleic acid (LA) compared to their wild-type (WT) counterparts, while no such differences were observed in male Ghsr-/- and WT mice. Overall, our results are consistent with the interpretation that ghrelin in the taste system is involved in the complex sensing and recognition of fat compounds. Ghrelin-GHS-R signaling may play a critical role in the recognition of fatty acids in female mice, and this differential regulation may contribute to their distinct ingestive behaviors.
Collapse
Affiliation(s)
- Ashley N. Calder
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA; (A.N.C.); (N.S.D.)
| | - Tian Yu
- Department of Cell & Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Naima S. Dahir
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA; (A.N.C.); (N.S.D.)
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA;
| | - Timothy A. Gilbertson
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Correspondence: ; Tel.: +1-321-266-7245
| |
Collapse
|
40
|
Lacerda DC, Urquiza-Martínez MV, Manhaes-de-Castro R, Visco DB, Derosier C, Mercado-Camargo R, Torner L, Toscano AE, Guzmán-Quevedo O. Metabolic and neurological consequences of the treatment with polyphenols: a systematic review in rodent models of noncommunicable diseases. Nutr Neurosci 2021; 25:1680-1696. [PMID: 33650943 DOI: 10.1080/1028415x.2021.1891614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND Noncommunicable diseases (NCDs) lead to drastic metabolic alterations with associated energy balance and body weight changes, two related physiological processes regulated by the brain. Polyphenol-based treatments for NCDs have emerged as a promising therapy, which seems to involve the energy balance modulation. However, it remains unclear what the most effective polyphenols-based treatment is to attenuate adverse effects in the energy balance of NCDs. OBJECTIVES This systematic review aimed to evaluate the literature on the metabolic and neurological effects of polyphenols-based treatment in rodent models of NCDs. METHODS Literature search was carried out in the following databases: CINAHL, Medline/PubMed, SCOPUS, and Web of Science. For title and abstract screening, original papers with polyphenols exposure in rodents were selected. For full-text screening, studies with models of NCDs that reported metabolic and neurological outcomes when treated with polyphenols were selected for inclusion in this review. RESULTS 23 articles, using individual compound (11 articles) or polyphenols extracts (12 articles), were included in this review: 5 articles using tea polyphenols, 12 articles using grape-derived polyphenols, 3 articles using the polyphenol quercetin, and 3 articles using other polyphenol sources. Most results agree on the beneficial effect of polyphenols in attenuating alterations in energy balance and body weight. Such effects were associated with neuroprotective responses in different brain areas including hippocampus and hypothalamus. CONCLUSION In conclusion, this review shows that the treatment with polyphenols, especially resveratrol or quercetin, attenuates the adverse effects of NCDs on energy balance and are associated with neuroprotective effects.
Collapse
Affiliation(s)
- D C Lacerda
- Unidade de Estudos em Nutrição e Plasticidade Fenotípica do Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil.,Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil
| | - M V Urquiza-Martínez
- Facultad de Químico-Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, México.,Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - R Manhaes-de-Castro
- Unidade de Estudos em Nutrição e Plasticidade Fenotípica do Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil.,Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil
| | - D B Visco
- Unidade de Estudos em Nutrição e Plasticidade Fenotípica do Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil.,Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil
| | - C Derosier
- Pós-Graduação em Neuropsiquiatria e Ciências do Comportamento, Universidade Federal de Pernambuco, Recife, Brazil
| | - R Mercado-Camargo
- Facultad de Químico-Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, México
| | - L Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - A E Toscano
- Unidade de Estudos em Nutrição e Plasticidade Fenotípica do Departamento de Nutrição, Universidade Federal de Pernambuco, Recife, Brazil.,Pós-Graduação em Neuropsiquiatria e Ciências do Comportamento, Universidade Federal de Pernambuco, Recife, Brazil.,Departmento de Enfermagem, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil
| | - O Guzmán-Quevedo
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico.,Pós-Graduação em Neuropsiquiatria e Ciências do Comportamento, Universidade Federal de Pernambuco, Recife, Brazil.,Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Mexico
| |
Collapse
|
41
|
Cornejo MP, Mustafá ER, Cassano D, Banères JL, Raingo J, Perello M. The ups and downs of growth hormone secretagogue receptor signaling. FEBS J 2021; 288:7213-7229. [PMID: 33460513 DOI: 10.1111/febs.15718] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
The growth hormone secretagogue receptor (GHSR) has emerged as one of the most fascinating molecules from the perspective of neuroendocrine control. GHSR is mainly expressed in the pituitary and the brain, and plays key roles regulating not only growth hormone secretion but also food intake, adiposity, body weight, glucose homeostasis and other complex functions. Quite atypically, GHSR signaling displays a basal constitutive activity that can be up- or downregulated by two digestive system-derived hormones: the octanoylated-peptide ghrelin and the liver-expressed antimicrobial peptide 2 (LEAP2), which was recently recognized as an endogenous GHSR ligand. The existence of two ligands with contrary actions indicates that GHSR activity can be tightly regulated and that the receptor displays the capability to integrate such opposing inputs in order to provide a balanced intracellular signal. This article provides a summary of the current understanding of the biology of ghrelin, LEAP2 and GHSR and discusses the reconceptualization of the cellular and physiological implications of the ligand-regulated GHSR signaling, based on the latest findings.
Collapse
Affiliation(s)
- María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Jean-Louis Banères
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Ecole Nationale Supérieure de Chimie de Montpellier, Faculté de Pharmacie, Montpellier cedex 5, France
| | - Jesica Raingo
- Laboratory of Electrophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], Buenos Aires, Argentina
| |
Collapse
|
42
|
Khelifa MS, Skov LJ, Holst B. Biased Ghrelin Receptor Signaling and the Dopaminergic System as Potential Targets for Metabolic and Psychological Symptoms of Anorexia Nervosa. Front Endocrinol (Lausanne) 2021; 12:734547. [PMID: 34646236 PMCID: PMC8503187 DOI: 10.3389/fendo.2021.734547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Anorexia Nervosa (AN) is a complex disease that impairs the metabolic, mental and physiological health of affected individuals in a severe and sometimes lethal way. Many of the common symptoms in AN patients, such as reduced food intake, anxiety, impaired gut motility or overexercising are connected to both the orexigenic gut hormone ghrelin and the dopaminergic system. Targeting the ghrelin receptor (GhrR) to treat AN seems a promising possibility in current research. However, GhrR signaling is highly complex. First, the GhrR can activate four known intracellular pathways Gαq, Gαi/o, Gα12/13 and the recruitment of β-arrestin. Biased signaling provides the possibility to activate or inhibit only one or a subset of the intracellular pathways of a pleiotropic receptor. This allows specific targeting of physiological functions without adverse effects. Currently little is known on how biased signaling could specifically modulate GhrR effects. Second, GhrR signaling has been shown to be interconnected with the dopaminergic system, particularly in the context of AN symptoms. This review highlights that a biased agonist for the GhrR may be a promising target for the treatment of AN, however extensive and systematic translational studies are still needed and the connection to the dopaminergic system has to be taken into account.
Collapse
|
43
|
Zhang C, Yuan J, Lin Q, Li M, Wang L, Wang R, Chen X, Jiang Z, Zhu K, Chang X, Wang B, Dong J. Ghrelin in the lateral parabrachial nucleus influences the excitability of glucosensing neurons, increases food intake and body weight. Endocr Connect 2020; 9:1168-1177. [PMID: 33112816 PMCID: PMC7774750 DOI: 10.1530/ec-20-0285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/13/2020] [Indexed: 01/10/2023]
Abstract
Ghrelin plays a pivotal role in the regulation of food intake, body weight and energy metabolism. However, these effects of ghrelin in the lateral parabrachial nucleus (LPBN) are unexplored. C57BL/6J mice and GHSR-/- mice were implanted with cannula above the right LPBN and ghrelin was microinjected via the cannula to investigate effect of ghrelin in the LPBN. In vivo electrophysiological technique was used to record LPBN glucose-sensitive neurons to explore potential udnderlying mechanisms. Microinjection of ghrelin in LPBN significantly increased food intake in the first 3 h, while such effect was blocked by [D-Lys3]-GHRP-6 and abolished in GHSR-/- mice. LPBN ghrelin microinjection also significantly increased the firing rate of glucose-excited (GE) neurons and decreased the firing rate of glucose-inhibited (GI) neurons. Additionally, LPBN ghrelin microinjection also significantly increased c-fos expression. Chronic ghrelin administration in the LPBN resulted in significantly increased body weight gain. Meanwhile, no significant changes were observed in both mRNA and protein expression levels of UCP-1 in BAT. These results demonstrated that microinjection of ghrelin in LPBN could increase food intake through the interaction with growth hormone secretagogue receptor (GHSR) in C57BL/6J mice, and its chronic administration could also increase body weight gain. These effects might be associated with altered firing rate in the GE and GI neurons.
Collapse
Affiliation(s)
- Caishun Zhang
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Junhua Yuan
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Qian Lin
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Manwen Li
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Liuxin Wang
- Hyperbaric Oxygen Therapy Department, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Rui Wang
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Xi Chen
- Physiology Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhengyao Jiang
- Physiology Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Kun Zhu
- Intensive Care Unit Department, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoli Chang
- Institute of Acupuncture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bin Wang
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, China
- Medical Microbiology Department, College of Basic Medicine, Qingdao University, Qingdao, China
| | - Jing Dong
- Special Medicine Department, College of Basic Medicine, Qingdao University, Qingdao, China
- Physiology Department, College of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
44
|
"A LEAP 2 conclusions? Targeting the ghrelin system to treat obesity and diabetes". Mol Metab 2020; 46:101128. [PMID: 33246141 PMCID: PMC8085568 DOI: 10.1016/j.molmet.2020.101128] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The hormone ghrelin stimulates food intake, promotes adiposity, increases body weight, and elevates blood glucose. Consequently, alterations in plasma ghrelin levels and the functioning of other components of the broader ghrelin system have been proposed as potential contributors to obesity and diabetes. Furthermore, targeting the ghrelin system has been proposed as a novel therapeutic strategy for obesity and diabetes. SCOPE OF REVIEW The current review focuses on the potential for targeting ghrelin and other proteins comprising the ghrelin system as a treatment for obesity and diabetes. The main components of the ghrelin system are introduced. Data supporting a role for the endogenous ghrelin system in the development of obesity and diabetes along with data that seemingly refute such a role are outlined. An argument for further research into the development of ghrelin system-targeted therapeutic agents is delineated. Also, an evidence-based discussion of potential factors and contexts that might influence the efficacy of this class of therapeutics is provided. MAJOR CONCLUSIONS It would not be a "leap to" conclusions to suggest that agents which target the ghrelin system - including those that lower acyl-ghrelin levels, raise LEAP2 levels, block GHSR activity, and/or raise desacyl-ghrelin signaling - could represent efficacious novel treatments for obesity and diabetes.
Collapse
|
45
|
Pape J, Herbison AE, Leeners B. Recovery of menses after functional hypothalamic amenorrhoea: if, when and why. Hum Reprod Update 2020; 27:130-153. [PMID: 33067637 DOI: 10.1093/humupd/dmaa032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/12/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Prolonged amenorrhoea occurs as a consequence of functional hypothalamic amenorrhoea (FHA) which is most often induced by weight loss, vigorous exercise or emotional stress. Unfortunately, removal of these triggers does not always result in the return of menses. The prevalence and conditions underlying the timing of return of menses vary strongly and some women report amenorrhoea several years after having achieved and maintained normal weight and/or energy balance. A better understanding of these factors would also allow improved counselling in the context of infertility. Although BMI, percentage body fat and hormonal parameters are known to be involved in the initiation of the menstrual cycle, their role in the physiology of return of menses is currently poorly understood. We summarise here the current knowledge on the epidemiology and physiology of return of menses. OBJECTIVE AND RATIONALE The aim of this review was to provide an overview of (i) factors determining the recovery of menses and its timing, (ii) how such factors may exert their physiological effects and (iii) whether there are useful therapeutic options to induce recovery. SEARCH METHODS We searched articles published in English, French or German language containing keywords related to return of menses after FHA published in PubMed between 1966 and February 2020. Manuscripts reporting data on either the epidemiology or the physiology of recovery of menses were included and bibliographies were reviewed for further relevant literature. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) criteria served to assess quality of observational studies. OUTCOMES Few studies investigate return of menses and most of them have serious qualitative and methodological limitations. These include (i) the lack of precise definitions for FHA or resumption of menses, (ii) the use of short observation periods with unsatisfactory descriptions and (iii) the inclusion of poorly characterised small study groups. The comparison of studies is further hampered by very inhomogeneous study designs. Consequently, the exact prevalence of resumption of menses after FHA is unknown. Also, the timepoint of return of menses varies strongly and reliable prediction models are lacking. While weight, body fat and energy availability are associated with the return of menses, psychological factors also have a strong impact on the menstrual cycle and on behaviour known to increase the risk of FHA. Drug therapies with metreleptin or naltrexone might represent further opportunities to increase the chances of return of menses, but these require further evaluation. WIDER IMPLICATIONS Although knowledge on the physiology of return of menses is presently rudimentary, the available data indicate the importance of BMI/weight (gain), energy balance and mental health. The physiological processes and genetics underlying the impact of these factors on the return of menses require further research. Larger prospective studies are necessary to identify clinical parameters for accurate prediction of return of menses as well as reliable therapeutic options.
Collapse
Affiliation(s)
- J Pape
- Department of Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - A E Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge CB2 3EG, UK
| | - B Leeners
- Department of Reproductive Endocrinology, University Hospital Zurich, 8091 Zurich, Switzerland.,University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
46
|
Liu H, Luo J, Guillory B, Chen JA, Zang P, Yoeli JK, Hernandez Y, Lee IIG, Anderson B, Storie M, Tewnion A, Garcia JM. Ghrelin ameliorates tumor-induced adipose tissue atrophy and inflammation via Ghrelin receptor-dependent and -independent pathways. Oncotarget 2020; 11:3286-3302. [PMID: 32934774 PMCID: PMC7476735 DOI: 10.18632/oncotarget.27705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue (AT) atrophy is a hallmark of cancer cachexia contributing to increased morbidity/mortality. Ghrelin has been proposed as a treatment for cancer cachexia partly by preventing AT atrophy. However, the mechanisms mediating ghrelin's effects are incompletely understood, including the extent to which its only known receptor, GHSR-1a, is required for these effects. This study characterizes the pathways involved in AT atrophy in the Lewis Lung Carcinoma (LLC)-induced cachexia model and those mediating the effects of ghrelin in Ghsr +/+ and Ghsr -/- mice. We show that LLC causes AT atrophy by inducing anorexia, and increasing lipolysis, AT inflammation, thermogenesis and energy expenditure. These changes were greater in Ghsr -/-. Ghrelin administration prevented LLC-induced anorexia only in Ghsr +/+, but prevented WAT lipolysis, inflammation and atrophy in both genotypes, although its effects were greater in Ghsr +/+. LLC-induced increases in BAT inflammation, WAT and BAT thermogenesis, and energy expenditure were not affected by ghrelin. In conclusion, ghrelin ameliorates WAT inflammation, fat atrophy and anorexia in LLC-induced cachexia. GHSR-1a is required for ghrelin's orexigenic effect but not for its anti-inflammatory or fat-sparing effects.
Collapse
Affiliation(s)
- Haiming Liu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Gerontology and Geriatric Medicine, University of Washington Department of Medicine, Seattle, WA, USA.,These authors contributed equally to this work
| | - Jiaohua Luo
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Environmental Hygiene, College of Preventive Medicine, Army Medical University, Chongqing, China.,These authors contributed equally to this work
| | - Bobby Guillory
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ji-An Chen
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Health Education, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Pu Zang
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Endocrinology, Nanjing Jinling Hospital, Nanjing, China
| | - Jordan K Yoeli
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yamileth Hernandez
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ian In-Gi Lee
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Gerontology and Geriatric Medicine, University of Washington Department of Medicine, Seattle, WA, USA
| | - Barbara Anderson
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Gerontology and Geriatric Medicine, University of Washington Department of Medicine, Seattle, WA, USA
| | - Mackenzie Storie
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Gerontology and Geriatric Medicine, University of Washington Department of Medicine, Seattle, WA, USA
| | - Alison Tewnion
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Jose M Garcia
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Gerontology and Geriatric Medicine, University of Washington Department of Medicine, Seattle, WA, USA.,Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
47
|
Cornejo MP, Barrile F, Cassano D, Aguggia JP, García Romero G, Reynaldo M, Andreoli MF, De Francesco PN, Perello M. Growth hormone secretagogue receptor in dopamine neurons controls appetitive and consummatory behaviors towards high-fat diet in ad-libitum fed mice. Psychoneuroendocrinology 2020; 119:104718. [PMID: 32535402 DOI: 10.1016/j.psyneuen.2020.104718] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/10/2023]
Abstract
Growth hormone secretagogue receptor (GHSR), the receptor for ghrelin, is expressed in key brain nuclei that regulate food intake. The dopamine (DA) pathways have long been recognized to play key roles mediating GHSR effects on feeding behaviors. Here, we aimed to determine the role of GHSR in DA neurons controlling appetitive and consummatory behaviors towards high fat (HF) diet. For this purpose, we crossed reactivable GHSR-deficient mice with DA transporter (DAT)-Cre mice, which express Cre recombinase under the DAT promoter that is active exclusively in DA neurons, to generate mice with GHSR expression limited to DA neurons (DAT-GHSR mice). We found that DAT-GHSR mice show an increase of c-Fos levels in brain areas containing DA neurons after ghrelin treatment, in a similar fashion as seen in wild-type mice; however, they did not increase food intake or locomotor activity in response to systemically- or centrally-administered ghrelin. In addition, we found that satiated DAT-GHSR mice displayed both anticipatory activity to scheduled HF diet exposure and HF intake in a binge-like eating protocol similar to those in wild-type mice, whereas GHSR-deficient mice displayed impaired responses. We conclude that GHSR expression in DA neurons is sufficient to both mediate increased anticipatory activity to a scheduled HF diet exposure and fully orchestrate binge-like HF intake, but it is insufficient to restore the acute orexigenic or locomotor effects of ghrelin treatment. Thus, GHSR in DA neurons affects appetitive and consummatory behaviors towards HF diet that take place in the absence of caloric needs.
Collapse
Affiliation(s)
- María Paula Cornejo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Franco Barrile
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Julieta Paola Aguggia
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Guadalupe García Romero
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Mirta Reynaldo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - María Florencia Andreoli
- Laboratory of Experimental Neurodevelopment, Institute of Development and Pediatric Research (IDIP), La Plata Children's Hospital and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)], La Plata, Buenos Aires, Argentina
| | - Pablo Nicolás De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina.
| |
Collapse
|
48
|
Mani BK, Puzziferri N, He Z, Rodriguez JA, Osborne-Lawrence S, Metzger NP, Chhina N, Gaylinn B, Thorner MO, Thomas EL, Bell JD, Williams KW, Goldstone AP, Zigman JM. LEAP2 changes with body mass and food intake in humans and mice. J Clin Invest 2020; 129:3909-3923. [PMID: 31424424 DOI: 10.1172/jci125332] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Acyl-ghrelin administration increases food intake, body weight, and blood glucose. In contrast, mice lacking ghrelin or ghrelin receptors (GHSRs) exhibit life-threatening hypoglycemia during starvation-like conditions, but do not consistently exhibit overt metabolic phenotypes when given ad libitum food access. These results, and findings of ghrelin resistance in obese states, imply nutritional state dependence of ghrelin's metabolic actions. Here, we hypothesized that liver-enriched antimicrobial peptide-2 (LEAP2), a recently characterized endogenous GHSR antagonist, blunts ghrelin action during obese states and postprandially. To test this hypothesis, we determined changes in plasma LEAP2 and acyl-ghrelin due to fasting, eating, obesity, Roux-en-Y gastric bypass (RYGB), vertical sleeve gastrectomy (VSG), oral glucose administration, and type 1 diabetes mellitus (T1DM) using humans and/or mice. Our results suggest that plasma LEAP2 is regulated by metabolic status: its levels increased with body mass and blood glucose and decreased with fasting, RYGB, and in postprandial states following VSG. These changes were mostly opposite of those of acyl-ghrelin. Furthermore, using electrophysiology, we showed that LEAP2 both hyperpolarizes and prevents acyl-ghrelin from activating arcuate NPY neurons. We predict that the plasma LEAP2/acyl-ghrelin molar ratio may be a key determinant modulating acyl-ghrelin activity in response to body mass, feeding status, and blood glucose.
Collapse
Affiliation(s)
- Bharath K Mani
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Nancy Puzziferri
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA.,Department of Surgery, Veterans Administration North Texas Heath Care System, Dallas, Texas, USA
| | | | - Juan A Rodriguez
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Sherri Osborne-Lawrence
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Nathan P Metzger
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Navpreet Chhina
- PsychoNeuroEndocrinology Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, and.,Computational, Cognitive and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Bruce Gaylinn
- Department of Endocrinology, University of Virginia, Charlottesville, Virginia, USA
| | - Michael O Thorner
- Department of Endocrinology, University of Virginia, Charlottesville, Virginia, USA
| | - E Louise Thomas
- Research Centre for Optimal Health, University of Westminster, London, United Kingdom
| | - Jimmy D Bell
- Research Centre for Optimal Health, University of Westminster, London, United Kingdom
| | | | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, and.,Computational, Cognitive and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Jeffrey M Zigman
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| |
Collapse
|
49
|
Abstract
Ghrelin is a key signal driving energy seeking and storage in order to reverse energy deficit. In line with this view, the metabolic status of an organism predicts sensitivity to ghrelin, with fasting increasing and obesity decreasing ghrelin sensitivity. However, the mechanism responsible for controlling this sensitivity is unknown. In this issue of the JCI, Mani and colleagues show that plasma levels of plasma liver-enriched antimicrobial peptide-2 (LEAP2), a recently identified hormone that antagonizes the ghrelin receptor, are inversely correlated with those of plasma acyl-ghrelin under conditions of both energy deficit and energy surplus in mice and humans. Their results show that a fall in plasma LEAP2 during energy deficit facilitates the actions of acyl-ghrelin, whereas increased LEAP2 in obesity suppresses the actions of acyl-ghrelin. This important discovery helps reshape our understanding of ghrelin function and may provide a new approach to aiding weight maintenance after diet-induced weight loss.
Collapse
|
50
|
Sovetkina A, Nadir R, Fung JNM, Nadjarpour A, Beddoe B. The Physiological Role of Ghrelin in the Regulation of Energy and Glucose Homeostasis. Cureus 2020; 12:e7941. [PMID: 32499981 PMCID: PMC7266561 DOI: 10.7759/cureus.7941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ghrelin is a peptide hormone that is primarily released from the stomach. It is best known for its role in appetite initiation. However, evidence also suggests that ghrelin may play a much wider role in energy homeostasis and glucose metabolism. It is known that exogenous ghrelin exerts an orexigenic signal via growth hormone secretagogue receptors in the arcuate nucleus of the hypothalamus. However, blocking ghrelin signalling in the arcuate nucleus does not decrease feeding. Evidence now proposes that an alternative pathway for ghrelin’s action is via the vagus nerve. Furthermore, it has been suggested that ghrelin signalling is an important physiological regulator of body adiposity and energy storage. Ghrelin also seems to be important in controlling glucose metabolism through action in the pancreatic islets of Langerhans, representing a promising novel therapeutic target in diabetes treatment. Despite these findings, further research in humans is required before ghrelin can be indicated as a therapeutic target in obesity or diabetes. This review summarises the current literature concerning ghrelin’s physiological roles in energy and glucose homeostasis.
Collapse
Affiliation(s)
| | - Rans Nadir
- Faculty of Medicine, Imperial College London, London, GBR
| | | | | | | |
Collapse
|