1
|
Yada T, Dezaki K, Iwasaki Y. GLP-1 and ghrelin inversely regulate insulin secretion and action in pancreatic islets, vagal afferents, and hypothalamus for controlling glycemia and feeding. Am J Physiol Cell Physiol 2025; 328:C1793-C1807. [PMID: 40241252 DOI: 10.1152/ajpcell.00168.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025]
Abstract
Glucagon-like peptide-1 (GLP-1) was discovered as an incretin hormone, which is released from the intestine upon nutrient intake and stimulates insulin secretion from the pancreatic islet β-cells. Subsequently, its ability to suppress appetite was recognized. Ghrelin, discovered as the ligand for growth hormone secretagogue-receptor (GHS-R), is released from the stomach and produces appetite. Later, its ability to inhibit insulin secretion and elevate blood glucose was found. Thus, GLP-1 and ghrelin regulate insulin secretion and appetite toward opposite directions. The receptor agonists for GLP-1 and ghrelin have been developed and are now used to treat metabolic diseases, in which insulin plays a key role. However, underlying action mechanism and possible interplay of these hormones have remained elusive. Here, we describe that GLP-1 and ghrelin reciprocally regulate the insulin system. GLP-1 enhances and ghrelin suppresses insulin secretion in pancreatic β-cells. Moreover, GLP-1 cooperates with and ghrelin counteracts insulin action in the vagal afferent and hypothalamic arcuate nucleus (ARC) neurons, the interfaces between the peripheral metabolism and brain. Notably, ghrelin rises and works preprandially and GLP-1 rises and works postprandially. The interplay of ghrelin, GLP-1, and insulin leads to optimal circadian control of feeding, glycemia, and metabolism.
Collapse
Affiliation(s)
- Toshihiko Yada
- Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Osaka, Japan
- Department of Diabetes, Endocrinology and Metabolism/Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Gifu, Japan
- Division of Integrative Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan
| | - Katsuya Dezaki
- Department of Physiology, Faculty of Pharmacy, Iryo Sosei University, Iwaki, Japan
- Division of Integrative Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
- Division of Integrative Physiology, Jichi Medical University School of Medicine, Shimotsuke, Japan
| |
Collapse
|
2
|
Wu YC, Beets I, Fox BW, Fajardo Palomino D, Chen L, Liao CP, Vandewyer E, Lin LY, He CW, Chen LT, Lin CT, Schroeder FC, Pan CL. Intercellular sphingolipid signaling mediates aversive learning in C. elegans. Curr Biol 2025; 35:2323-2336.e9. [PMID: 40252647 DOI: 10.1016/j.cub.2025.03.082] [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: 12/10/2024] [Revised: 03/02/2025] [Accepted: 03/31/2025] [Indexed: 04/21/2025]
Abstract
Physiological stress in non-neural tissues drives aversive learning for sensory cues associated with stress. However, the identities of signals derived from non-neural tissues and the mechanisms by which these signals mediate aversive learning remain elusive. Here, we show that intercellular sphingolipid signaling contributes to aversive learning under mitochondrial stress in C. elegans. We found that stress-induced aversive learning requires sphingosine kinase, SPHK-1, the enzyme that produces sphingosine-1-phosphate (S1P). Genetic and biochemical studies revealed an intercellular signaling pathway in which intestinal or hypodermal SPHK-1 signals through the neuronal G protein-coupled receptor, SPHR-1, and modulates responses of the octopaminergic RIC neuron to promote aversive learning. We further show that SPHK-1-mediated sphingolipid signaling is required for learned aversion of Chryseobacterium indologenes, a bacterial pathogen found in the natural habitats of C. elegans, which causes mitochondrial stress. Taken together, our work reveals a sphingolipid signaling pathway that communicates from intestinal or hypodermal tissues to neurons to promote aversive learning in response to mitochondrial stress and pathogen infection.
Collapse
Affiliation(s)
- Yu-Chun Wu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Isabel Beets
- Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, Isabel Beets, Leuven, Belgium
| | - Bennett William Fox
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, 533 Tower Road, Ithaca, NY 14853, USA
| | - Diana Fajardo Palomino
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, 533 Tower Road, Ithaca, NY 14853, USA
| | - Li Chen
- Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, Isabel Beets, Leuven, Belgium
| | - Chien-Po Liao
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Elke Vandewyer
- Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, Isabel Beets, Leuven, Belgium
| | - Liang-Yi Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chun-Wei He
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Li-Tzu Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chih-Ta Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Frank C Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, 533 Tower Road, Ithaca, NY 14853, USA
| | - Chun-Liang Pan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan.
| |
Collapse
|
3
|
Franchini L, Porter JJ, Lueck JD, Orlandi C. G zESTY as an optimized cell-based assay for initial steps in GPCR deorphanization. Nat Commun 2025; 16:4521. [PMID: 40374633 PMCID: PMC12081699 DOI: 10.1038/s41467-025-59850-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are key pharmacological targets, yet many remain underutilized due to unknown activation mechanisms and ligands. Orphan GPCRs, lacking identified natural ligands, are a high priority for research, as identifying their ligands will aid in understanding their functions and potential as drug targets. Most GPCRs, including orphans, couple to Gi/o/z family members, however current assays to detect their activation are limited, hindering ligand identification efforts. We introduce GzESTY, a sensitive, cell-based assay developed in an easily deliverable format designed to study the pharmacology of Gi/o/z-coupled GPCRs and assist in deorphanization. We optimized assay conditions and developed an all-in-one vector employing cloning methods to ensure the correct expression ratio of GzESTY components. GzESTY successfully assessed activation of a library of ligand-activated GPCRs, detecting both full and partial agonism, and responses from endogenous GPCRs. Notably, with GzESTY we established the presence of endogenous ligands for GPR176 and GPR37 in brain extracts, validating its use in deorphanization efforts. This assay enhances the ability to find ligands for orphan GPCRs, expanding the toolkit for GPCR pharmacologists.
Collapse
Grants
- R01DC022104 U.S. Department of Health & Human Services | NIH | National Institute on Deafness and Other Communication Disorders (NIDCD)
- R01HL153988 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL153988 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | NIH | National Institute on Deafness and Other Communication Disorders (NIDCD)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- This work was supported by start-up funding from the Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry to C.O.; Ernest J. Del Monte Institute for Neuroscience Pilot Program, University of Rochester, to C.O.; University Research Award, University of Rochester to C.O; NIDCD/NIH grant R01DC022104 to C.O.; R01HL153988 to J.D.L.; this work was aided by the GCE4All Biomedical Technology Optimization and Dissemination Center supported by National Institute of General Medical Science grant RM1-GM144227; The Foundation Blanceflor Boncompagni Ludovisi-née Bildt fellowship to L.F.
Collapse
Affiliation(s)
- Luca Franchini
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Joseph J Porter
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - John D Lueck
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
4
|
Liu Y, Xu B, Cheng S, Wang Y, Ding J, Shen X, Wu B, Xu L, Wei J. Novel chimeric peptides of endomorphin-2 and the active fragments of ghrelin exhibit blood-brain barrier permeability and central antinociceptive effects with reduced opioid-related side effects. Neuropharmacology 2025; 269:110324. [PMID: 39904410 DOI: 10.1016/j.neuropharm.2025.110324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Pharmacological research has showed that multi-targeted drug therapies offer superior efficacy and reduced side effects compared to single-target drug therapies. In this study, we designed and characterized four novel chimeric peptides G (1-5)-EM2, EM2-G (1-5), G (1-9)-EM2 and EM2-G (1-9) which incorporate endomorphin-2 (EM-2) and the active fragments of ghrelin. Calcium mobilization assays revealed that these four chimeric peptides acted as weak mixed agonists for the μ-opioid receptor (MOR), κ-opioid receptor (KOR), and growth hormone secretagogue receptor 1α (GHS-R1α). The results of fluorescence imaging experiments indicated that G (1-5)-EM2 and G (1-9)-EM2 could penetrate the blood-brain barrier (BBB) following intravenous (i.v.) injection. All chimeric peptides induced almost equal antinociceptive effects compared with EM-2 or better antinociceptive effects than EM-2 after intracerebroventricular (i.c.v.) injection in the acute pain in mice. Among them, G (1-5)-EM2 could cross the BBB and enter the brain to induce antinociceptive effect through central opioid receptors after i. v. Injection. Our findings demonstrated that the chimeric peptides produced significant antinociception mainly via MOR, DOR and GHS-R1α without inducing antinociceptive tolerance, or with a lower tendency for antinociceptive tolerance after i. c.v. Injection in the acute pain in mice. Furthermore, the chimeric peptides mitigated or eliminated the digestive side effects associated with EM-2. The collective results highlight G (1-5)-EM2 as the most promising candidate among the chimeric peptides. The chimeric peptides represent a promising class of potential analgesics for clinical pain management. However, further optimization is necessary to maximize their therapeutic potential.
Collapse
Affiliation(s)
- Yongling Liu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province, 730000, PR China
| | - Songxia Cheng
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Yan Wang
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Jiali Ding
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Xiaoyu Shen
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Bing Wu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Liangquan Xu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China
| | - Jie Wei
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, PR China; Jiangxi Province Key Laboratory of Brain Science and Brian Health, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
5
|
Sumimoto T, Tanaka R, Sato A, Tatsuta R, Itoh H. Wide-range and high-throughput quantification of anamorelin in human plasma using ultra-high-performance liquid chromatography coupled to tandem mass spectrometry. Clin Biochem 2025; 138:110949. [PMID: 40373991 DOI: 10.1016/j.clinbiochem.2025.110949] [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: 01/20/2025] [Revised: 04/27/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
OBJECTIVE Cancer cachexia is characterized by weight loss, muscle mass loss, and reduced food intake. Anamorelin is a ghrelin receptor agonist approved for the treatment of cancer cachexia. In this study, we established and validated an assay for quantification of anamorelin in human plasma. METHODS For quantification of anamorelin, samples were pretreated with solid-phase extraction and analyzed by ultra-high performance liquid chromatography coupled to tandem mass spectrometry (UHPLC-MS/MS). This analytical method was validated in accordance with the Food and Drug Administration (FDA) bioanalytical method validation guidance. We used the established assay to quantify plasma anamorelin concentrations in five patients with cancer cachexia treated with anamorelin. RESULTS The validation results of this assay method met the acceptance criteria recommended by the FDA guidance. Within-batch and batch-to-batch precision at the lower limit of quantification and three quality control levels were within 6.20 % and 6.55 % coefficient of variation, respectively. Within-batch and batch-to-batch accuracies ranged from -2.58 to -1.33 % and -3.78 to -1.69 %, respectively. Recovery rates and matrix effects corrected by internal standard were 82.7-84.2 % and 102.7-104.6 %, respectively. Using the established assay with a calibration range of 0.1-2500 ng/mL, plasma anamorelin concentrations were successfully quantified in all 15 plasma samples from 5 patients with cancer cachexia. CONCLUSIONS We established and validated a method to measure plasma anamorelin concentrations using UHPLC/MS-MS combined with SPE, and successfully applied the novel method to measure plasma anamorelin concentrations in patients with cancer cachexia. By measuring plasma anamorelin concentrations in large scale studies, the established quantitative method is expected to contribute to the pharmacokinetic study of anamorelin.
Collapse
Affiliation(s)
- Takahiro Sumimoto
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan.
| | - Ryota Tanaka
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Ayaka Sato
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Ryosuke Tatsuta
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Hiroki Itoh
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| |
Collapse
|
6
|
Wilms JN, Hendriks S, Sugino T, Ghaffari MH, Steele MA, Sauerwein H, Martín-Tereso J, Leal LN. Inclusion of a spray-dried fat concentrate containing tributyrin and tricaproin in milk replacer enhanced increased feed intake, growth, and elicited metabolic and endocrine responses in ad libitum-fed calves. J Dairy Sci 2025:S0022-0302(25)00321-2. [PMID: 40349754 DOI: 10.3168/jds.2025-26331] [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: 01/17/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Fat composition of milk replacer (MR) for calves differs from that of bovine milk fat, resulting in lower levels of butyric (C4:0) and caproic acid (C6:0). These fatty acids play a critical role in the gastrointestinal and metabolic development of calves by supporting rumen epithelial growth, enhancing energy metabolism, and promoting overall gut health. The objective of this study was to evaluate the effects of inclusion of a spray-dried fat concentrate containing tributyrin (TB) and tricaproin (TC) in a high-fat MR on growth, feed intake, and metabolic profiles of ad libitum-fed calves. Forty-eight newborn Holstein calves were blocked based on arrival sequence. Within each block of 2 calves, calves were randomly assigned to a control MR (CON, n = 24) including vegetable fats from palm, coconut, and linseed fats, or to an experimental MR including the same fat blend, to which TB and TC (TRI, n = 24) was added to the same levels found in milk fat. Both MR contained 23.7% crude protein, 27.7% fat, and 35.6% lactose (DM basis) and were fed at 13.5% solids. Calves were group housed and fed ad libitum MR with automated feeders. Weaning was gradual and induced between wk 7-10, after which calves were only fed solid feeds. Starter feed, chopped straw, and water were offered ad libitum throughout the whole study period. Calves were weighed and blood was collected once weekly at 1300h. Calves fed TRI showed a ∼50% reduction in therapeutic intervention days compared with CON. In addition, calves fed TRI consumed significantly more MR and starter feed, resulting in a greater growth. Serum NEFA and plasma total cholesterol were lower, whereas the enzymatic activity of serum ALP was higher in calves fed TRI than in CON. In addition, calves fed TRI had lower serum ghrelin and higher insulin-like growth factor-I concentrations. Incorporating TB and TC is a suitable strategy to increase solid feed intake upon weaning, resulting in better growth performance in ad libitum systems, and to improve health of dairy rearing calves.
Collapse
Affiliation(s)
- J N Wilms
- Trouw Nutrition Research and Development, P.O. Box 299, 3800 AG, Amersfoort, the Netherlands; Department of Animal Bioscience, University of Guelph, Guelph, ON, Canada.
| | - S Hendriks
- Trouw Nutrition Research and Development, P.O. Box 299, 3800 AG, Amersfoort, the Netherlands; Adaptation Physiology, Wageningen University, P.O. Box 338, 6700 AH, Wageningen, the Netherlands
| | - T Sugino
- The Research Center for Animal Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan 739-8528
| | - M H Ghaffari
- Institute of Animal Science, University of Bonn, 53111 Bonn, Germany
| | - M A Steele
- Department of Animal Bioscience, University of Guelph, Guelph, ON, Canada
| | - H Sauerwein
- Institute of Animal Science, University of Bonn, 53111 Bonn, Germany
| | - J Martín-Tereso
- Trouw Nutrition Research and Development, P.O. Box 299, 3800 AG, Amersfoort, the Netherlands
| | - L N Leal
- Trouw Nutrition Research and Development, P.O. Box 299, 3800 AG, Amersfoort, the Netherlands
| |
Collapse
|
7
|
Hao H, Bao F, Wang Y, Li N, Gong Y. Peptide therapy: new promising therapeutics for acute kidney injury. Drug Discov Today 2025; 30:104377. [PMID: 40348078 DOI: 10.1016/j.drudis.2025.104377] [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: 12/24/2024] [Revised: 04/01/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Acute kidney injury (AKI) is a common fatal condition among hospitalized patients. AKI may be induced by a variety of complicating factors such as sepsis, ischemia-reperfusion injury, nephrotoxic substances, and rhabdomyolysis. At present, symptomatic treatment is mainly used, and there are no US Food and Drug Administration (FDA)-approved drugs for the prevention or treatment of AKI. Peptides have become a promising area of research in AKI treatment because of their high efficiency and low toxicity. In this paper, we systematically review the experimental advancements of peptide therapy for AKI, analyze the mechanism of peptide action in different pathological models, discuss the challenges facing peptide therapy, and provide a scientific basis for further clinical research.
Collapse
Affiliation(s)
- Herui Hao
- School of Disaster and Emergency Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Faculty of Medicine, Tianjin University, China
| | - Fengjiao Bao
- School of Disaster and Emergency Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Faculty of Medicine, Tianjin University, China
| | - Yuru Wang
- School of Disaster and Emergency Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Faculty of Medicine, Tianjin University, China
| | - Ning Li
- School of Disaster and Emergency Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Faculty of Medicine, Tianjin University, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China.
| | - Yanhua Gong
- School of Disaster and Emergency Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Faculty of Medicine, Tianjin University, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China.
| |
Collapse
|
8
|
Ma J, Zhang J, Liu J, Zhao J, Wang X, Li Z, Lv T, Zhang Y. Ghrelin/GHSR system attenuates collagen-induced arthritis in mice and ameliorates inflammation in human rheumatoid arthritis fibroblast-like synoviocytes. Biochem Pharmacol 2025; 238:116973. [PMID: 40339721 DOI: 10.1016/j.bcp.2025.116973] [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: 12/27/2024] [Revised: 04/12/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Ghrelin, an acylated peptide hormone, acts through its sole known receptor, the growth hormone secretagogue receptor (GHSR).Previous research indicated that ghrelin may be involved in rheumatoid arthritis (RA), yet the specific mechanisms remain unclear. This study aimed to explore the mechanism of ghrelin in RA synovial inflammation. Serum and synovial tissue from RA patients were collected for ghrelin expression analysis. We conducted our study using a collagen-induced arthritis (CIA) mouse model and an in vitro model using fibroblast-like synoviocytes (FLSs) induced by tumor necrosis factor-alpha (TNF-α). RNA-sequencing was performed to identify the potential signaling pathways involved in RA. Ghsr shRNA interference was used to assess whether the ghrelin receptor was involved. Ghrelin expression was decreased in synovial tissue of RA patients, and was negatively associated with TNF-α in the synovial fluid. In vivo experiments, acyl-ghrelin effectively suppressed CIA development, and Ghsr-/- mice exhibited the significantly aggravated arthritis symptoms of CIA mice. RNA sequence analyses of synovial tissue in Ghsr-/- and wild type mice indicated that ghrelin/GHSRsystem may inhibit inflammation through the PI3K/AKT pathway. In RA-FLSs, we found that acyl-ghrelin significantly suppressed the TNF-α induced increase in p-PI3K, p-AKT, p-NF-κB p65, IL-6 and IL-1β in RA FLSs. The effects of acyl-ghrelin on inflammatory factors were attenuated by the PI3K/AKT agonists. Ghsr shRNA reversed the anti-inflammatory effects of acyl-ghrelin. These results indicated that ghrelin/GHSR system has an important role in RA and could be a suitable candidate for RA therapy.
Collapse
Affiliation(s)
- Junxian Ma
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China; Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China
| | - Jinshan Zhang
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China; Department of Basic Medical Morphology, Medical College, Xijing University, 1 of Xijing Road, Xi'an, China
| | - Jie Liu
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China
| | - Jie Zhao
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China
| | - Xia Wang
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China
| | - Zhen Li
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China.
| | - Tingting Lv
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China.
| | - Yan Zhang
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China.
| |
Collapse
|
9
|
Ahn B, Wanagat J, Cleary C, Ainsworth HC, Kim E, Kim H. Unacylated Ghrelin Counteracts Contractile and Mitochondrial Dysfunction in Cancer Cachexia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.29.649515. [PMID: 40376088 PMCID: PMC12080946 DOI: 10.1101/2025.04.29.649515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Background Cancer cachexia is a complex metabolic syndrome that severely impacts patient mobility, treatment strategies, and quality of life. However, no treatments are available to mitigate the debilitating consequences of cancer cachexia. Unacylated ghrelin (UnAG), the main circulating form of ghrelin, enhances muscle growth and mitochondrial function in various diseases, but its effects in cancer cachexia remain to be tested. Methods Male C57Bl6/N mice were assigned to one of three treatment groups: non-tumor-bearing (NTB), tumor-bearing (TB), or tumor-bearing treated with unacylated ghrelin (TB+UnAG). Over four weeks, we monitored body weight, food intake, and tumor size. We assessed muscle mass, contractility, mitochondrial oxygen consumption rate (OCR), and reactive oxygen species (ROS) production. Proteomic analysis was performed to elucidate the downstream effects of UnAG. Cell culture assays were performed to measure the in vitro effects of cancer cell-secreted factors and UnAG on myoblasts. Results Gastrocnemius and quadriceps muscle masses were reduced by 20-30% in TB mice compared to NTB controls; however, UnAG treatment prevented approximately 50% of this loss. Beyond muscle mass, UnAG enhanced the isometric maximum specific force of the extensor digitorum longus by 70% in TB mice. This improvement in muscle quality was associated with preferential upregulation of myosin heavy chain expression in TB+UnAG mice. UnAG also increased mitochondrial OCR while reducing ROS production. Mitochondrial DNA (mtDNA) copy number, which was reduced in TB mice, was restored by UnAG, while the reduced mtDNA mutation frequency in TB mice was maintained with treatment, indicating improved mtDNA integrity. Consistent with enhanced mitochondrial function, treadmill running time was significantly increased in TB+UnAG mice. Proteomic analysis revealed that UnAG downregulated proteins associated with proteolysis, while normalizing antioxidant enzyme thioredoxin and proteins involved in calcium handling. Cancer cell-conditioned medium reduced myotube width in vitro, but UnAG treatment preserved myotube structure.. Conclusion UnAG protects against cancer cachexia by targeting multiple risk factors, including myosin heavy chain expression, mitochondrial bioenergetics, and modulation of protein degradation pathways.
Collapse
|
10
|
Hiki N, Higuchi T, Kumagai K, Okuno K, Minoura H, Sato Y, Fujita S, Harada H, Chuman M, Washio M, Sakuraya M, Niihara M, Kumamoto Y, Naitoh T, Yamashita K. Appetite-preserving gastrectomy (APG) for esophagogastric junction cancer: preserving the residual stomach as an endocrine organ. Gastric Cancer 2025; 28:527-536. [PMID: 40100486 PMCID: PMC11993504 DOI: 10.1007/s10120-025-01603-z] [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: 12/23/2024] [Accepted: 02/20/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Loss of appetite following gastric cancer surgery, particularly total gastrectomy, significantly impacts patient quality of life due to the removal of the ghrelin-secreting region. We developed appetite-preserving gastrectomy (APG), a modified total gastrectomy that preserves this region. METHODS Ten consecutive patients with esophagogastric junction cancer who were indicated for total gastrectomy and underwent APG between April 2023 and April 2024 were evaluated for early surgical outcomes, appetite, and changes in weight and body composition. RESULTS There were no postoperative complications of grade II or higher (Clavien-Dindo classification). Appetite, assessed using the Simplified Nutritional Appetite Questionnaire, showed no significant impairment at 3 months (14.5 points, P = 0.82) and 6 months (15 points, P = 0.44) postoperatively compared with preoperative values. Oral calorie intake was maintained at 3 months (1675 kcal, P = 0.97) and 6 months (1675 kcal, P = 0.22) postoperatively compared with preoperative levels. The patients' body weight decreased by 9.2% at 6 months postoperatively compared with preoperative values, but their lean body mass remained stable. Although a significant decrease in the blood Ghrelin levels was observed postoperatively, 53% and 60.4% of the preoperative levels was maintained at one month and 6 months, respectively. CONCLUSIONS APG is a safe procedure that preserves the residual stomach as an endocrine organ, maintains ghrelin secretion and appetite, and prevents muscle loss. However, further trials are required to compare the efficacy of APG with total gastrectomy in preventing postoperative appetite loss.
Collapse
Affiliation(s)
- Naoki Hiki
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Tadashi Higuchi
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Koshi Kumagai
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Kota Okuno
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hiroyuki Minoura
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yumi Sato
- Department of Nutrition, Kitasato University Hospital, Sagamihara, Japan
| | - Shohei Fujita
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hiroki Harada
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Motohiro Chuman
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Marie Washio
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Mikiko Sakuraya
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Masahiro Niihara
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yusuke Kumamoto
- Department of General-Pediatric Hepato Biliary Pancreatic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takeshi Naitoh
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Keishi Yamashita
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
11
|
Zhang C, Liu Y, Gao Y, Li M, Dong Y, Liu X, Li J. The sex-specific relationship of ghrelin and cognition in Chinese han first-episode drug-naive major depressive disorder. J Neural Transm (Vienna) 2025; 132:699-707. [PMID: 39849211 DOI: 10.1007/s00702-025-02880-9] [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: 12/03/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
In major depressive disorder (MDD), alterations in ghrelin levels and cognitive impairment coexist, yet their association has remained largely elusive. This study aimed to investigate the association between ghrelin levels and cognition in both MDD patients and healthy controls (HCs) while also exploring sex-specific differences in this correlation. A total of 155 Chinese Han subjects, including 90 first-episode drug-naive MDD patients and 65 HCs, were enrolled. Ghrelin levels were measured using ELISA kits, and neurocognitive assessments were conducted using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). MDD patients exhibited significantly higher ghrelin levels and lower cognitive scores of RBANS compared to HCs. However, there was no significant correlation between ghrelin levels and cognitive function in both MDD patients and HCs. Exploratory analyses revealed sex-specific associations between ghrelin and cognitive function, particularly in MDD patients. Females with MDD showed distinct patterns of association between ghrelin levels and cognitive domains such as attention and language, which were not observed in healthy controls or male MDD patients. The relationship between ghrelin and cognition only existed in MDD patients, not in the HCs, and there was a sex-specific difference in this association. Further research on the mechanism of ghrelin in the cognitive function of MDD should focus on sex differences.
Collapse
Affiliation(s)
- Chuhao Zhang
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Yuan Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Ying Gao
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Meijuan Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Yeqing Dong
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Xueying Liu
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China.
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China.
| |
Collapse
|
12
|
Alvarez-Crespo M, Gil-Lozano M, Diz-Chaves Y, González-Matias LC, Mallo F. Elevation of ghrelin by B-adrenergic activation is independent of glucose variations and feeding regimen in the rat. Endocrine 2025; 88:434-445. [PMID: 40169505 PMCID: PMC12069131 DOI: 10.1007/s12020-024-04156-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/28/2024] [Indexed: 04/03/2025]
Abstract
Ghrelin is a signal involved in the initiation of meals in rodents and humans. Circulating ghrelin levels are elevated before mealwes and reduced after food intake. Several factors have been identified as effective modulators of ghrelin levels. Vagal activation reduced ghrelin in rats, as well as oral carbohydrate and lipid administration in rats and humans. Some hormones, such as incretins, also reduce ghrelin: GLP-1 reduced ghrelin in humans, and Ex4, a GLP-1 receptor agonist, potently inhibited ghrelin in rodents. On the other hand, fasting promotes increases in ghrelin that anticipate the start of meals. We report that beta-adrenergic activation with isoproterenol promotes large acute elevations of circulating ghrelin levels, both in anesthetized and conscious freely-moving rats, either on "ad libitum" feeding or on a fasting regimen.These effects are dose-dependent, caused by intravenous, intraperitoneal, and oral administration, and independent of variations in glucose levels. Pharmacological modulation of β1 and β2 adrenergic receptors with specific agonists and antagonists showed that ghrelin increases are stimulated by β1-adrenergic activation, but also partially by β2-adrenergic activation, suggesting that activation of both is necessary to elicit complete ghrelin elevations. Meanwhile, glucose increases dependent on adrenergic activation appear to be mediated only by β2-adrenergic receptors. In addition, the effects of isoproterenol on increasing ghrelin levels are potent enough to overcome the marked inhibition exerted by exendin-4 that we have previously demonstrated. We also found that administration of isoproterenol in drinking water increases basal ghrelin levels and simultaneous food intake in animals eating ad libitum. Beta-adrenergic activation promotes increases in ghrelin levels in vivo prior to food intake, both in rats eating ad libitum and in fasting rats that already have elevated ghrelin levels, in a time- and dose-dependent manner. In addition, the effects of isoproterenol on increasing ghrelin levels are potent enough to overcome the marked inhibition exerted by exendin-4 that we have previously demonstrated. We also found that administration of isoproterenol in drinking water increases basal ghrelin levels and simultaneous food intake in animals eating ad libitum. Beta-adrenergic activation promotes increases in ghrelin levels in vivo prior to food intake, both in eating ad libitum and in fasting rats that already have elevated ghrelin levels, in a time- and dose-dependent manner.
Collapse
Affiliation(s)
- Mayte Alvarez-Crespo
- LabEndoTeam - Laboratory of Endocrinology - Department of Functional Biology and Health Sciences - University of Vigo - Campus as Lagoas - Marcosende, Vigo, Spain
| | - Manuel Gil-Lozano
- LabEndoTeam - Laboratory of Endocrinology - Department of Functional Biology and Health Sciences - University of Vigo - Campus as Lagoas - Marcosende, Vigo, Spain
| | - Yolanda Diz-Chaves
- LabEndoTeam - Laboratory of Endocrinology - Department of Functional Biology and Health Sciences - University of Vigo - Campus as Lagoas - Marcosende, Vigo, Spain
| | - Lucas Carmelo González-Matias
- LabEndoTeam - Laboratory of Endocrinology - Department of Functional Biology and Health Sciences - University of Vigo - Campus as Lagoas - Marcosende, Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur). SERGAS-UVIGO, Vigo, Spain
| | - Federico Mallo
- LabEndoTeam - Laboratory of Endocrinology - Department of Functional Biology and Health Sciences - University of Vigo - Campus as Lagoas - Marcosende, Vigo, Spain.
- Galicia Sur Health Research Institute (IIS Galicia Sur). SERGAS-UVIGO, Vigo, Spain.
| |
Collapse
|
13
|
Lufrano D, Gong C, Cecarini V, Cuccioloni M, Bonfili L, Sturaro C, Bettegazzi B, Ruzza C, Perelló M, Angeletti M, Eleuteri AM. An Insight into Neuronal Processing of Ghrelin: Effects of a Bioactive Ghrelin Derivative on Proteolytic Pathways and Mitophagy. Mol Neurobiol 2025:10.1007/s12035-025-04976-5. [PMID: 40285938 DOI: 10.1007/s12035-025-04976-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Protein homeostasis (proteostasis) is preserved by an orchestrated network of molecular mechanisms that regulate protein synthesis, folding, and degradation, ensuring cellular integrity and function. Proteostasis declines with age and is related to pathologies such as neurodegenerative diseases and cardiac disorders, which are accompanied by the accumulation of toxic protein aggregates. In this context, therapeutic strategies enhancing the two primary degradative systems involved in the cellular clearance of those abnormal proteins, namely ubiquitin-proteasome system and autophagy-lysosomal pathway, represent a promising approach to counteract the collapse of proteostasis in such pathological conditions. In this work, we explored the processing of ghrelin, a pleiotropic peptide hormone linked to energy metabolism and higher brain functions, which is reported to modulate the protein degradative mechanisms. According to our data, ghrelin is processed by serine hydrolases secreted into the conditioned medium of SH-SY5Y neuroblastoma cell line, commonly used in neurotoxicology and neuroscience research. Ghrelin processing leads to the formation of a shorter peptide (ghrelin(1-11)) that stimulates both the cell proteasome system and autophagy-lysosomal pathway, encompassing the selective autophagy of mitochondria. Our findings suggest that ghrelin processing may contribute to the maintenance of neuronal proteostasis.
Collapse
Affiliation(s)
- Daniela Lufrano
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
- Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional La Plata (UNLP), CONICET, B1900 AVW, La Plata, Argentina.
| | - Chunmei Gong
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education, Hainan Medical University, 571199, Haikou, China
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Massimiliano Cuccioloni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Chiara Sturaro
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | | | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | - Mario Perelló
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de La Provincia de Buenos Aires (CIC-PBA), B1906 APM, La Plata, Argentina
| | - Mauro Angeletti
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
| |
Collapse
|
14
|
Zhang M, Yang L, Mi X, Hu G, Lu Y, Wang C, Yang J, Sun X, Niu M, Li X, Wang S, Zhang J, Yu H, Wang Y, Yu M, Li N, Zhou Y. GHS-R1a signaling drives anxiety-related behavior by shaping excitability of ventromedial hypothalamic neurons. Nat Commun 2025; 16:3858. [PMID: 40274845 PMCID: PMC12022087 DOI: 10.1038/s41467-025-59116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
The neural substrates of anxiety are poorly understood, which hinders treatment of anxiety disorders. Here we found, αCaMKII+ neurons in the ventral medial hypothalamic nucleus (VMH) responded to stressors with increased activity in male mice, both under physiological conditions and after repeated restraint stress. Activation of VMH αCaMKII+ neurons were necessary and sufficient to ameliorate stress-induced anxiety. The peripheral metabolic hormone ghrelin and receptor GHS-R1a play a complex role in emotion regulation; however, the mechanism is uncertain. A delayed increase in GHS-R1a expression in VMH αCaMKII+ neurons coincided with the development of stress-induced enhancement of anxiety-related behavior. GHS-R1a expression in VMH αCaMKII+ neurons promoted anxiety-related behavior, whereas GHS-R1a knockdown had the opposite effect. GHS-R1a upregulation inhibited the excitability of VMH αCaMKII+ neurons. We conclude that GHSR1a signaling drives stress-induced anxiety by shaping the activity of VMH αCaMKII+ neurons. GHS-R1a may be a therapeutic target for treating anxiety disorders such as post-traumatic stress disorder.
Collapse
Affiliation(s)
- Meng Zhang
- School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266113, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
- College of Agriculture and Bioengineering, Heze University, Heze, Shandong, 274000, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Xue Mi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Gonghui Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yingchang Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Chen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Jie Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
- Dongying No.1 Middle School, Dongying, Shandong, 257000, China
| | - Xiaomin Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
- Qingdao West Coast New Area No. 9 Senior High School, Qingdao, Shandong, 266500, China
| | - Minglu Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
- Qingdao Endocrine diabetes Hospital, Qingdao, Shandong, 266000, China
| | - Xianchao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Sihan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Jingsai Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Hanbing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yuyang Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Ming Yu
- School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266113, China
| | - Nan Li
- School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266113, China
| | - Yu Zhou
- School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266113, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao, Shandong, 266071, China.
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong, 266071, China.
- Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| |
Collapse
|
15
|
Punt LD, Kooijman S, Mutsters NJM, Yue K, van der Kaay DCM, van Tellingen V, Bakker-van Waarde WM, Boot AM, van den Akker ELT, van Boekholt AA, de Groote K, Kruijsen AR, van Nieuwaal-van Maren NHG, Woltering MC, Heijligers M, van der Heyden JC, Bannink EMN, Rinne T, Hannema SE, de Waal WJ, Delemarre LC, Rensen PCN, de Bruin C, van Duyvenvoorde HA, Visser JA, Delhanty PJD, Losekoot M, Wit JM, Joustra SD. Loss-of-Function GHSR Variants Are Associated With Short Stature and Low IGF-I. J Clin Endocrinol Metab 2025; 110:e1303-e1314. [PMID: 39785833 PMCID: PMC12012706 DOI: 10.1210/clinem/dgaf010] [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: 10/25/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/12/2025]
Abstract
CONTEXT The growth hormone (GH) secretagogue receptor, encoded by GHSR, is expressed on somatotrophs of the pituitary gland. Stimulation with its ligand ghrelin, as well as its constitutive activity, enhances GH secretion. Studies in knockout mice suggest that heterozygous loss-of-function of GHSR is associated with decreased GH response to fasting, but patient observations in small case reports have been equivocal. OBJECTIVE This work aims to establish the phenotype of GHSR haploinsufficiency and its growth response to GH treatment. METHODS This case series includes 26 patients with short stature and heterozygous GHSR variants. Pathogenicity was studied in vitro using total protein levels, cell surface expression, and receptor activity in basal, stimulated, and inhibited states. RESULTS Ten different variants were identified, of which 6 were novel. Variants showed either partial or complete loss of function, primarily through loss of constitutive activity. Patients (aged 4.0-15.1 years) had proportionate short stature (height -2.8 ± 0.5 SDS), failure to thrive with low appetite (n = 4), a mean serum insulin-like growth factor-I (IGF-I) of -1.6 ± 0.7 SDS, and a normal stimulated GH response. Nine patients received GH treatment, showing a height gain of 0.9 ± 0.4 SDS after 1 year and 1.5 ± 0.4 SDS after 2 years (n = 5). CONCLUSION This study combines phenotypical and functional data in a uniquely large group of children with short stature carrying GHSR variants, and shows their good response to GH treatment. The results strengthen the hypothesis of GHSR's role in GH secretion.
Collapse
Affiliation(s)
- Lauren D Punt
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Noa J M Mutsters
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Kaiming Yue
- Division of Endocrinology, Department of Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Daniëlle C M van der Kaay
- Division of Pediatric Endocrinology, Department of Pediatrics, Erasmus University Medical Centre, Sophia Children's Hospital, 3015 GD Rotterdam, the Netherlands
| | - Vera van Tellingen
- Department of Pediatrics, Catharina Hospital, 5623 EJ Eindhoven, the Netherlands
| | - Willie M Bakker-van Waarde
- Division of Pediatric Endocrinology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Annemiek M Boot
- Division of Pediatric Endocrinology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Erica L T van den Akker
- Division of Pediatric Endocrinology, Department of Pediatrics, Erasmus University Medical Centre, Sophia Children's Hospital, 3015 GD Rotterdam, the Netherlands
| | | | - Kirsten de Groote
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Anne R Kruijsen
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | | | - M Claire Woltering
- Department of Pediatrics, Reinier de Graaf Gasthuis, 2625 AD Delft, the Netherlands
| | - Malou Heijligers
- Department of Clinical Genetics, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands
| | - Josine C van der Heyden
- Department of Pediatrics, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, the Netherlands
| | - Ellen M N Bannink
- Department of Pediatrics, Tergooi MC, 1212 VG Hilversum, the Netherlands
| | - Tuula Rinne
- Department of Human Genetics, Radboud UMC, 6525 GA Nijmegen, the Netherlands
| | - Sabine E Hannema
- Department of Pediatric Endocrinology, Amsterdam UMC, location Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1081 HV Amsterdam, the Netherlands
- Amsterdam Reproduction and Development, 1105 AZ Amsterdam, the Netherlands
| | - Wouter J de Waal
- Department of Pediatrics, Diakonessenhuis, 3582 KE Utrecht, the Netherlands
| | - Lucia C Delemarre
- Department of Pediatrics, Amstelland Hospital, 1186 AM Amstelveen, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Christiaan de Bruin
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | | | - Jenny A Visser
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Patric J D Delhanty
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Monique Losekoot
- Department of Clinical Genetics, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Jan M Wit
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Sjoerd D Joustra
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
16
|
Smith KR, Schreyer CC, Bello NT, Goodman E, Tamashiro KLK, Moran TH, Guarda AS. Blunted cold pressor test-induced cortisol but not total ghrelin response in women with bulimia nervosa following a standardized sweet-fat liquid meal. Appetite 2025; 213:108020. [PMID: 40268247 DOI: 10.1016/j.appet.2025.108020] [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: 02/01/2024] [Revised: 04/11/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Bulimia nervosa (BN) involves recurrent episodes of uncontrolled eating followed by compensatory behaviors. Stress is implicated in BN, affecting the hypothalamic-pituitary-adrenal (HPA) axis and ghrelin, a key appetite-regulating hormone. Studies report conflicting HPA axis findings in BN, necessitating further investigation. OBJECTIVE To examine the impact of acute stress on cortisol and serum ghrelin and eating disorder symptoms in women with BN and healthy controls (HC). METHODS Participants underwent a socially evaluated cold pressor test (CPT) and control condition (quiet rest) before consuming a sweet-fat liquid meal (530 Kcal milkshake). Hormonal responses and subjective measures of stress, interoception, and appetite were assessed. RESULTS In BN but not HC, desire to binge remained consistently high in both conditions and correlated with perceived hunger. There were no group differences in total ghrelin levels and levels were not influenced by the CPT. Baseline cortisol levels were similar for HC and BN groups, however BN subjects did not demonstrate a CPT-induced elevation in cortisol as observed in HC. CONCLUSION Results confirm HPA axis dysregulation in BN in response to a passive stressor and liquid meal challenge. Meal-related total ghrelin however does not appear to be involved in the stress response in women with or without BN. Desire to binge is persistent in BN, irrespective of the presence or absence of an acute stressor with a sweet-fat liquid meal and may be associated with heightened emotional states in general.
Collapse
Affiliation(s)
- Kimberly R Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Colleen C Schreyer
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nicholas T Bello
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Ethan Goodman
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Angela S Guarda
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Ida T, Matsui K, Nagata S, Nakamachi T, Shiimura Y, Sato T, Kojima M. Discovery of Feeding Regulatory Peptides and The Importance of Peptide Discovery Research. Kurume Med J 2025:MS7134001. [PMID: 40254448 DOI: 10.2739/kurumemedj.ms7134001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Bioactive peptides consist of multiple linked amino acids that are secreted from cells and act on specific receptors in order to transmit information from one cell to another. Through signal transduction, bioactive peptides regulate various physiological functions in the body, and the discovery of new bioactive peptides is therefore likely to lead to the development of various diagnostic and therapeutic agents. In this article, we have focused on the bioactive peptides that are known as feeding regulatory peptides. They are among the bioactive peptides discovered as ligands for G protein-coupled receptors (GPCRs), and we have reviewed their diverse functions. In addition, the status of structural analysis of GPCRs, which is necessary in the drug discovery process, and research on orphan GPCRs, for which new ligands are expected to be discovered in the future, is introduced to systematize modern peptide research and discuss future developments in bioactive peptide research.
Collapse
Affiliation(s)
- Takanori Ida
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki
| | - Kazuma Matsui
- Division of Molecular Genetics, Institute of Life Science, Kurume University
| | - Sayaka Nagata
- Department of Food Science and Technology, Faculty of Health and Nutrition, Minami Kyushu University
| | - Tomoya Nakamachi
- Laboratory of Regulatory Biology, Faculty of Science, Academic Assembly, University of Toyama
| | - Yuki Shiimura
- Division of Molecular Genetics, Institute of Life Science, Kurume University
- Department of Cell Biology, Graduate School of Medicine, Kyoto University
| | - Takahiro Sato
- Division of Molecular Genetics, Institute of Life Science, Kurume University
| | - Masayasu Kojima
- Division of Molecular Genetics, Institute of Life Science, Kurume University
| |
Collapse
|
18
|
Oruk S, Ergul Erkec O, Huyut Z, Acikgoz E. Neuroprotective effects of ghrelin in cuprizone-induced rat model of multiple sclerosis. Metab Brain Dis 2025; 40:176. [PMID: 40214860 PMCID: PMC11991981 DOI: 10.1007/s11011-025-01603-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Multiple sclerosis (MS) is an inflammatory central nervous system disease characterized by demyelination and axonal loss and is the main cause of non-traumatic neurological disability in young adults. Although there are several treatment approaches to manage the disease, there is no definitive cure for multiple sclerosis. Inflammation and oxidative stress are known to play important roles in the pathophysiology of MS. Ghrelin, a peptide secreted by the stomach, is reported to have neuroprotective properties through several pathways, including attenuating oxidative stress and inflammation. In the present study cuprizone (CPZ)-induced model of MS was used in Wistar albino rats to study the possible anti-inflammatory, antioxidant and neuroprotective effects of ghrelin. Rats were randomly divided into six groups: Control groups (Control35 and Control-S42), demyelination group, remyelination group, remyelination + ghrelin (20 µg/kg) group and remyelination + ghrelin (40 µg/kg) group. Y maze test was performed on the rats on their last day of the experiment. Oxidative stress and inflammatory parameters were investigated in brain using commercial kits by enzyme-linked immunosorbent assay (ELISA). Luxol fast blue (LFB) and hematoxylen&eosin (H&E) staining were performed in brain tissues. CPZ leads to a significant decrease in glutathione peroxidase (GSH-Px) levels and myelin content and a significant increase in malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-ɑ), interleukin- 6 (IL- 6) levels, the number of lymphatic cells and inflammatory cells. A significant increase in the antioxidant parameter levels and a significant decrease in MDA levels were found in the ghrelin treated groups (p < 0.05). CPZ leads to irregular, fragmented, demyelinating nerve fibers. A more significant remyelination was observed in the ghrelin treated groups compared to the other groups (p < 0.05). In conclusion, ghrelin treatment showed neuroprotective and antioxidant properties and reduced demyelination in the CPZ-induced rat model of multiple sclerosis.
Collapse
Affiliation(s)
- Sezai Oruk
- Department of Medical Physiology, Institute of Health Sciences, Van Yuzuncu Yil University, Van, Turkey
| | - Ozlem Ergul Erkec
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey.
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
19
|
Shiimura Y, Kojima M, Sato T. How the ghrelin receptor recognizes the acyl-modified orexigenic hormone. Front Mol Neurosci 2025; 18:1549366. [PMID: 40260011 PMCID: PMC12009760 DOI: 10.3389/fnmol.2025.1549366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
Ghrelin, discovered in 1999 as an endogenous ligand of the growth hormone secretagogue receptor (now known as the ghrelin receptor), is a peptide hormone with diverse physiological activities, such as stimulation of growth hormone release, increased appetite, fat accumulation, thermoregulation, and cardioprotection. As a distinctive feature, ghrelin needs to undergo octanoylation, a specific acyl modification, to exert its biological activities. Although the ghrelin receptor specifically recognizes this modification, the underlying molecular mechanism had remained unclear for decades. Recent advancements in structural biology have facilitated the elucidation of this recognition mechanism 25 years after ghrelin's discovery. This review highlights the structural basis of ghrelin octanoylation, particularly emphasizing the mechanism by which the ghrelin receptor recognizes this acyl-modified hormone.
Collapse
Affiliation(s)
- Yuki Shiimura
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masayasu Kojima
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Takahiro Sato
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| |
Collapse
|
20
|
Anderson KC, Grammer EE, Stephenson B, Stahl ME, Weeldreyer NR, Liu Z, Love KM, Allen JD, Weltman A. The interrelationship among exercise intensity, endothelial function, and ghrelin in healthy humans. Physiol Rep 2025; 13:e70213. [PMID: 40214273 PMCID: PMC11987205 DOI: 10.14814/phy2.70213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 04/14/2025] Open
Abstract
Ghrelin circulates in acylated (AG) and deacylated (DAG) isoforms and both may impact endothelial function (EF). Although acute exercise has been shown to modulate ghrelin levels and EF, data on the impact of exercise intensity on these parameters are scarce. To investigate the effect of exercise intensity and sex on EF and ghrelin levels, nine males (age: 43.8 ± 10.3 y; BMI: 22.5 ± 1.8 kg/m2) and eight females (age: 33.75 ± 10.2 y; BMI: 22.4 ± 1.6 kg/m2) completed a maximal cycle ergometer lactate threshold (LT)/VO2peak test. This test determined the exercise intensity for three visits: (a) CON, no exercise; (b) MOD, the power output (PO) at LT; (c) HIGH, the PO associated with 75% of the difference between LT and VO2peak. Ghrelin levels and EF [flow-mediated dilation (FMD), shear rate (SR)] were measured at baseline and then 30-120 min post-exercise. HIGH and MOD increased FMD (p < 0.0001). Each ghrelin isoform was suppressed by HIGH; only females exhibited reduced DAG levels in HIGH compared to MOD and CON (p < 0.0001-0.004). FMD was associated with ghrelin levels in females (r = -0.26-0.47). High-intensity exercise is key for ghrelin suppression and appears to only be weakly/moderately related to EF.
Collapse
Affiliation(s)
- Kara C. Anderson
- Department of Kinesiology, School of Education and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of MedicineUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Emily E. Grammer
- Department of Kinesiology, School of Education and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Benjamin Stephenson
- Department of Kinesiology, School of Education and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Macy E. Stahl
- Department of Kinesiology, School of Education and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Nathan R. Weeldreyer
- Department of Kinesiology, School of Education and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Zhenqi Liu
- Department of MedicineUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Kaitlin M. Love
- Department of MedicineUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Jason D. Allen
- Department of Kinesiology, School of Education and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of MedicineUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Arthur Weltman
- Department of Kinesiology, School of Education and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of MedicineUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| |
Collapse
|
21
|
Kore MS, Mamsa R, Patil D, Bhatt LK. Ghrelin in Depression: A Promising Therapeutic Target. Mol Neurobiol 2025; 62:4237-4249. [PMID: 39424690 DOI: 10.1007/s12035-024-04554-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Depression is a widespread disease affecting over 300 million individuals of various ethnicities and socioeconomic backgrounds globally. It frequently strikes early in life and becomes a chronic or recurring lifelong illness. Out of the various hypotheses for the pathophysiology of depression, the gut-brain axis and stress hypothesis are the ones that need to be researched, as psychological stress impairs one or more pathways of the brain-gut axis and is likely to cause brain-gut axis dysfunction and depression. A dysfunctional reciprocal gut-brain relationship may contribute to many diseases, including inflammatory disorders, abnormal stress responses, impaired behavior, and metabolic changes. The hormone ghrelin is a topic of interest concerning the gut-brain axis as it interacts with the gut-brain axis indirectly via the central nervous system or via crossing the blood-brain barrier. Ghrelin release is also affected by the gut microbes, which has also been discussed in the review. This review elaborates on Ghrelin's role in depression and its effect on various aspects like neurogenesis, HPA axis, and neuroinflammation. Furthermore, this review focuses on ghrelin as a potential target for alleviation of depressive symptoms.
Collapse
Affiliation(s)
- Mikhil Santosh Kore
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Rumaiza Mamsa
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Dipti Patil
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
22
|
Sakai K, Nakazato Y, Shiimura Y, Zhang W, Nakazato M. Ghrelin-LEAP2 interactions along the stomach-liver axis. Endocr J 2025; 72:341-353. [PMID: 39756956 PMCID: PMC11997273 DOI: 10.1507/endocrj.ej24-0543] [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: 10/02/2024] [Accepted: 11/16/2024] [Indexed: 01/07/2025] Open
Abstract
Ghrelin produced in the stomach promotes food intake and GH secretion, and acts as an anabolic peptide during starvation. Ghrelin binds to the growth hormone secretagogue receptor, a G protein-coupled receptor (GPCR), whose high-resolution complex structures have been determined in the apo state and when bound to an antagonist. Anamorelin, a low-molecular-weight ghrelin agonist, has been launched in Japan for the treatment of cancer cachexia, and its therapeutic potential has attracted attention due to the various biological activities of ghrelin. In 2019, liver-expressed antimicrobial peptide (LEAP2), initially discovered as an antimicrobial peptide produced in the liver, was identified to be upregulated in the stomach of diet-induced obese mice after vertical sleeve gastrectomy. LEAP2 binds to the GHSR and antagonizes ghrelin's activities. The serum concentrations of human LEAP2 are positively correlated with body mass index, body fat accumulation, and fasting serum concentrations of glucose and triglyceride. Serum LEAP2 elevated and ghrelin reduced in obesity. Ghrelin and LEAP2 regulate body weight, food intake, and GH and blood glucose concentrations, and other physiological phenomena through their interactions with the same receptor, GHSR.
Collapse
Affiliation(s)
- Katsuya Sakai
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuki Nakazato
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuki Shiimura
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka 830-0011, Japan
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Weidong Zhang
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
- Laboratory of Biomolecular Analysis, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Masamitsu Nakazato
- Forefront Research Center, Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| |
Collapse
|
23
|
Zhang S, Kaiya H, Kitazawa T. Physiological roles of ghrelin in the regulation of gastrointestinal motility in vertebrates. Gen Comp Endocrinol 2025; 365:114698. [PMID: 40024446 DOI: 10.1016/j.ygcen.2025.114698] [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: 10/29/2024] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Ghrelin is known to be a multifunctional peptide hormone that stimulates not only growth hormone secretion and feeding but also gastrointestinal (GI) functions, including motility, secretion and mucosa proliferation. The aim of this review is to provide a comprehensive overview on the physiological roles of ghrelin in the regulation of GI motility from a comparative perspective. The effects of ghrelin on GI motility differ depending on the species, and ghrelin is a possible regulator of gastric migrating motor complexes (MMCs) in rodents, dogs and house musk shrew (suncus). However, the role of ghrelin has not been clarified in detail in other mammals, including humans and rabbits. Ghrelin is also effective to cause contraction in the GI tract of some non-mammals, but its physiological role is also not clarified at present. Distribution of the growth hormone secretagogue receptor (GHSR, ghrelin receptor) in the GI tract might be connected with the regulatory role of ghrelin in vertebrates. Comparative studies of ghrelin among animals and identification of knowledge gaps must lead us to the functional transition and importance of ghrelin in the GI tract.
Collapse
Affiliation(s)
- Shuangyi Zhang
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan; College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hiroyuki Kaiya
- Grandsoul Research Institute for Immunology, Inc., Uda, Nara 633-2221, Japan; Faculty of Science, University of Toyama, Toyama 930-8555, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
24
|
Bilgiç A, Cura M, Kılınç İ, Akça ÖF. Low Levels of Serum Ghrelin and Nesfatin-1 Are Associated With Anxiety Disorders in Children. Soa Chongsonyon Chongsin Uihak 2025; 36:69-77. [PMID: 40203141 PMCID: PMC11969048 DOI: 10.5765/jkacap.250001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Objectives Because appetite-regulating hormones are implicated in neuronal survival, growth, and differentiation, they have been suggested to play a role in anxiety disorders. To date, few studies have focused on the association between these hormones and anxiety disorders in children. This study investigated the potential differences in leptin, ghrelin, and nesfatin-1 serum levels in drug-naïve children with anxiety disorders, including social anxiety disorder, separation anxiety disorder, and generalized anxiety disorder, and in healthy controls. Methods This study included 45 children (14 boys and 31 girls) with anxiety disorders and 35 healthy controls (13 boys and 22 girls) aged 8-18 years. The severity of anxiety disorders and additional symptoms were evaluated using the Revised Child Anxiety and Depression Scales-Child Version. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate leptin, ghrelin, and nesfatin-1 serum levels. Results Leptin levels were significantly higher in children with anxiety disorders than in the control group, and ghrelin and nesfatin-1 levels were significantly lower in children with anxiety disorders than in the control group for girls and for the entire sample. However, only low nesfatin-1 levels were significantly associated with anxiety disorders in boys. In the entire sample, potential confounders such as age, sex, body mass index, and the severity of depressive symptoms were controlled for, and the results were the same for ghrelin and nesfatin-1 levels. However, the difference in leptin levels between groups was not significant. Conclusion These findings suggest that dysregulation of ghrelin and nesfatin-1 concentrations may be related to the etiopathogenesis of childhood anxiety disorders.
Collapse
Affiliation(s)
- Ayhan Bilgiç
- Department of Child and Adolescent Psychiatry, Faculty of
Medicine, Izmir University of Economics, Izmir,
Türkiye
| | - Merve Cura
- Department of Child and Adolescent Psychiatry, Etlik City
Hospital, Ankara, Türkiye
| | - İbrahim Kılınç
- Department of Biochemistry, Necmettin Erbakan University
Faculty of Medicine, Konya, Türkiye
| | - Ömer Faruk Akça
- Department of Child and Adolescent Psychiatry, Necmettin
Erbakan University Faculty of Medicine, Konya, Türkiye
| |
Collapse
|
25
|
Skoracka K, Hryhorowicz S, Schulz P, Zawada A, Ratajczak-Pawłowska AE, Rychter AM, Słomski R, Dobrowolska A, Krela-Kaźmierczak I. The role of leptin and ghrelin in the regulation of appetite in obesity. Peptides 2025; 186:171367. [PMID: 39983918 DOI: 10.1016/j.peptides.2025.171367] [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: 09/19/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
Leptin and ghrelin are two key hormones that play opposing roles in the regulation of appetite and energy balance. Ghrelin stimulates appetite and food intake following binding to receptors and the subsequent activation of orexigenic neurons in the arcuate nucleus. Leptin, conversely, has been demonstrated to suppress appetite and reduce food intake. This occurs through the inhibition of ghrelin-activated neurons, while simultaneously activating those that promote satiety and increase energy expenditure. A lack of biological response despite elevated leptin levels, which is known as leptin resistance, is observed in individuals with excess body weight and represents a significant challenge. As the dysregulation of ghrelin and leptin signalling has been linked to the development of obesity and other metabolic disorders, an in-depth understanding of the genetic determinants affecting these two hormones may facilitate a more comprehensive grasp of the intricate interactions that underpin the pathogenesis of obesity.
Collapse
Affiliation(s)
- Kinga Skoracka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewski 49, Poznan 60-355, Poland; Doctoral School, Poznan University of Medical Sciences, Bukowska 70, Poznan 60-812, Poland.
| | - Szymon Hryhorowicz
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, Poznan 60-479, Poland
| | - Piotr Schulz
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewski 49, Poznan 60-355, Poland
| | - Agnieszka Zawada
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewski 49, Poznan 60-355, Poland
| | - Alicja Ewa Ratajczak-Pawłowska
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewski 49, Poznan 60-355, Poland; Laboratory of Nutrigenetics, Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan 60-355, Poland
| | - Anna Maria Rychter
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewski 49, Poznan 60-355, Poland; Laboratory of Nutrigenetics, Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan 60-355, Poland
| | - Ryszard Słomski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, Poznan 60-479, Poland
| | - Agnieszka Dobrowolska
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewski 49, Poznan 60-355, Poland
| | - Iwona Krela-Kaźmierczak
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewski 49, Poznan 60-355, Poland; Laboratory of Nutrigenetics, Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Poznan 60-355, Poland.
| |
Collapse
|
26
|
Ergul Erkec O, Huyut Z, Acikgoz E, Huyut MT. Effects of exogenous ghrelin treatment on oxidative stress, inflammation and histological parameters in a fat-fed streptozotocin rat model. Arch Physiol Biochem 2025; 131:274-284. [PMID: 39324977 DOI: 10.1080/13813455.2024.2407551] [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: 05/16/2024] [Revised: 07/17/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
In this study, the anti-inflammatory, antioxidative, and protective effects of ghrelin were investigated in a fat-fed streptozotocin (STZ) rat model and compared with metformin, diabetes and the healthy control groups. Histopathological evaluations were performed on H&E-stained pancreas and brain sections. Biochemical parameters were investigated by enzyme-linked immunosorbent assay. Blood glucose levels were significantly decreased with ghrelin or metformin treatments than the diabetes group. STZ administration increased brain, renal and pancreatic IL-1β, TNF-α and MDA while decreasing GPX, CAT, SOD, and NGF levels. Ghrelin increased renal GPX, CAT, NGF pancreatic GPX, SOD, CAT, NGF and brain SOD, NGF while it decreased renal, pancreatic and brain IL-1β, TNF-α and MDA levels. Ghrelin reduced neuronal loss and degeneration in the cerebral cortex and hippocampus and greatly ameliorated diabetes-related damage in pancreas. In conclusion, the data suggested that ghrelin is an effective candidate as a protectant for reducing the adverse effects of diabetes.
Collapse
Affiliation(s)
- Ozlem Ergul Erkec
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Mehmet Tahir Huyut
- Department of Biostatistics, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| |
Collapse
|
27
|
Chang RC, Huang Y, To K, Whitlock RS, Nguyen KU, Joemon MC, Lopez M, Deeprompt KG, Shioda T, Blumberg B. Transgenerational Effects of the Obesogen Tributyltin on Metabolic Health in Mice: Interactions With a Western Diet. Endocrinology 2025; 166:bqaf063. [PMID: 40179257 PMCID: PMC11986328 DOI: 10.1210/endocr/bqaf063] [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: 11/04/2024] [Revised: 02/26/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Obesity is a global health crisis, with increasing evidence linking environmental factors such as exposure to endocrine-disrupting chemicals (EDCs) to its development. This study examines the transgenerational effects of exposure to the model obesogen, tributyltin (TBT), on obesity and metabolic health, specifically focusing on how these effects interact with a diet modeling the 50th percentile of US dietary consumption [the Total Western Diet (TWD)]. Pregnant F0 dams were exposed to TBT, and their offspring were subjected at adulthood to different diets, including a high-fat diet and TWD, across multiple subsequent generations (F1-F3). We found that TBT exposure predisposed male offspring to increased fat accumulation, insulin resistance, and metabolic dysfunction, effects that were exacerbated by the TWD. Notably, male offspring displayed elevated leptin levels, hepatic fibrosis, and inflammatory responses under TWD exposure, suggesting an additive or synergistic relationship between obesogen exposure and dietary fat intake. These transgenerational effects were largely absent in female offspring, underscoring sex-specific vulnerabilities to environmental and dietary factors. Our results demonstrated that the combination of prenatal TBT exposure and TWD amplifies metabolic disturbances across generations, highlighting the need to consider both environmental chemicals and dietary patterns in addressing the obesity pandemic. This study underscores the critical role of early-life EDC exposures and dietary factors in shaping long-term metabolic health and the potential for transgenerational programming of susceptibility to obesity and metabolic disorders.
Collapse
Affiliation(s)
- Richard C Chang
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Yikai Huang
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Kaitlin To
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Ryan Scott Whitlock
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Katelyn Uyen Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Michelle Clara Joemon
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Miranda Lopez
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Kritin Guy Deeprompt
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
| | - Toshi Shioda
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697-2300, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697-2300, USA
| |
Collapse
|
28
|
Xu ZY, Yu Y, Fu SX, Ma JY, Li BB. Effects of high-level ghrelin on intestinal epithelial cell proliferation, nutrient transport and intestinal mucosal immune barrier in chickens. Br Poult Sci 2025:1-16. [PMID: 40116599 DOI: 10.1080/00071668.2025.2456582] [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: 07/31/2024] [Accepted: 01/10/2025] [Indexed: 03/23/2025]
Abstract
1. Chicken ghrelin (GH) plays an important role in regulating growth hormone secretion, immunity and gastrointestinal motility. This study utilised haematoxylin-eosin staining, quantitative reverse transcription PCR and western blotting to examine the effects of high-level ghrelin on the proliferation of small intestinal epithelial cells, intestinal nutrient transport and the mucosal immune barrier in chicks.2. Eighty, 17-d-old layer type chicks were randomly divided into two groups: control (C treated with sterile phosphate buffer) and the ghrelin-treated group (GH; intraperitoneally injected with 0.5 nM GH per 100 g body weight). At 1, 3 and 5 d post-injection, six chicks from each group were randomly selected for sampling of the duodenum and ileum.3. Administering GH reduced the expression of proliferating cell nuclear antigen protein in the duodenum and leucine-rich repeat-containing G protein-coupled receptor 5 mRNA in both the duodenum and ileum. In addition, GH affected villus height and ratio of villus height to crypt (H/C) depth in these sections and fatty acid binding protein 6 expression in the ileum. The relative mRNA levels of oligopeptide transporter 1, solute carrier family 3 member 1, solute carrier family 1 member 1 and solute carrier family 5 member 1 were decreased by GH.4. Birds treated with GH had a decrease in duodenal intraepithelial lymphocytes, Paneth cells and ileal goblet cells. There was a reduction in mucin 2 mRNA in goblet cells and lysozyme C and phospholipaseA2 mRNA in Paneth cells. Additionally, the relative mRNA levels of avian β-defensin 1 (AvBD1), AvBD6 and AvBD7 in the duodenum and ileum decreased with GH administration.5. The GH inhibited proliferation of chicken duodenal epithelial cells and decreased surface area available for intestinal villus absorption. This affected the transport of intestinal amino acids, glucose and bile acids and impaired the function of the mucosal immune barrier in both the duodenum and ileum.
Collapse
Affiliation(s)
- Z-Y Xu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Y Yu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - S-X Fu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - J-Y Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - B-B Li
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| |
Collapse
|
29
|
Johansen VBI, Gradel AKJ, Holm SK, Cuenco J, Merrild C, Petersen N, Demozay D, Mani BK, Suppli MP, Grøndahl MFG, Lund AB, Knop FK, Prada-Medina CA, Hogendorf WFJ, Lykkesfeldt J, Merkestein M, Sakamoto K, Holst B, Clemmensen C. Regulation of LEAP2 by insulin and glucagon in mice and humans. Cell Rep Med 2025; 6:101996. [PMID: 40056903 PMCID: PMC11970398 DOI: 10.1016/j.xcrm.2025.101996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 09/14/2024] [Accepted: 02/10/2025] [Indexed: 03/10/2025]
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous antagonist and inverse agonist of the ghrelin receptor, countering ghrelin's effects on cell signaling and feeding. However, despite an emerging interest in LEAP2's physiology and pharmacology, its endocrine regulation remains unclear. Here, we report that plasma LEAP2 levels decrease significantly upon glucagon infusions during somatostatin clamps in humans. This effect is preserved in patients with obesity and type 2 diabetes while diminished following a hypercaloric diet and a sedentary lifestyle for 2 weeks. Additionally, insulin receptor antagonism offsets the upregulation of LEAP2 during the postprandial state in mice. Finally, insulin and glucagon receptor-expressing hepatocytes are the primary source of hepatic LEAP2 expression, coinciding with a putative enhancer-like signature bound by insulin- and glucagon-regulated transcription factors at the LEAP2 locus. Collectively, our findings implicate insulin and glucagon in regulating LEAP2 and warrant further investigations into the exact mechanisms orchestrating this endocrine axis.
Collapse
Affiliation(s)
- Valdemar Brimnes Ingemann Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Diabetes Pharmacology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark; Diabetes and Metabolism Biology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Anna Katrina Jógvansdóttir Gradel
- Diabetes Pharmacology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark; Section of Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephanie Kjærulff Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joyceline Cuenco
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Merrild
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Natalia Petersen
- Diabetes and Metabolism Biology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Damien Demozay
- Diabetes and Metabolism Biology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Bharath Kumar Mani
- Obesity and NASH Research, Global Drug Discovery, Novo Nordisk, Lexington, MA, USA
| | - Malte Palm Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Magnus F G Grøndahl
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Asger Bach Lund
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Filip Krag Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Cesar A Prada-Medina
- Systems Biology and Target Discovery, AI and Digital Research, Novo Nordisk Research Center Oxford, Novo Nordisk A/S, Oxford, UK
| | | | - Jens Lykkesfeldt
- Section of Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Myrte Merkestein
- Diabetes Pharmacology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
30
|
Pearl AJ, Maddern XJ, Pinares-Garcia P, Ursich LT, Anversa RG, Shesham A, Brown RM, Reed FM, Giardino WJ, Lawrence AJ, Walker LC. Midbrain ghrelin receptor signalling regulates binge drinking in a sex specific manner. Nat Commun 2025; 16:2568. [PMID: 40089486 PMCID: PMC11910522 DOI: 10.1038/s41467-025-57880-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Risky drinking rates are rising, particularly in women, yet sex as a biological variable has only recently gained traction. The centrally projecting Edinger-Westphal (EWcp) nucleus has emerged as a key regulator of alcohol consumption. Here we found that EWcppeptidergic cells reduce binge drinking specifically in female mice. We show this effect is mediated by the ghrelin receptor (GHSR), with EWcppeptidergic inhibition blocking ghrelin-induced drinking and Ghsr knockdown in EWcppeptidergic, but not EWcpglutamatergic or ventral tegmental area cells, reducing binge drinking in females, independent of circulating sex hormones. Female mice showed higher EWcp Ghsr expression, and EWcppeptidergic neurons were more sensitive to ghrelin. Moreover, intra-EWcp delivery of GHSR inverse agonist and antagonist reduced binge drinking, suggesting direct actions of ghrelin. These findings highlight the EWcp as a critical mediator of excessive alcohol consumption via GHSR in female mice, offering insights into the ghrelin system's role in alcohol consumption.
Collapse
Affiliation(s)
- Amy J Pearl
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Paulo Pinares-Garcia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Lauren T Ursich
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Roberta G Anversa
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Arnav Shesham
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Robyn M Brown
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Felicia M Reed
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - William J Giardino
- Dept. of Psychiatry and Behavioural Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
31
|
Zhu J, Zhou T, Chen G, Gao H, Chen X, Tuohetali A, Song Y, Pang D, Aimulajiang K. GHSR gene knockout alleviates the liver pathological response in Echinococcus granulosus infection by reducing parasite survival. Vet Res 2025; 56:55. [PMID: 40065480 PMCID: PMC11895129 DOI: 10.1186/s13567-025-01478-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/09/2025] [Indexed: 03/14/2025] Open
Abstract
Cystic echinococcosis (CE) is a parasitic disease caused by the larval stage of Echinococcus granulosus, and the immunosuppressive microenvironment exacerbates disease progression. Ghrelin, a peptide hormone, plays a role in modulating immune inflammation and may influence the progression of E. granulosus infection through its receptor, GHSR (growth hormone secretagogue receptor). However, whether GHSR downregulation can inhibit E. granulosus infection remains unclear. In this study, we extracted liver tissues from E. granulosus-infected mice and those treated with the GHSR antagonist [D-Lys3]-GHRP-6. Proteomic analysis revealed 341 differentially expressed proteins, of which 185 were upregulated and 156 were downregulated. Metabolomic sequencing revealed 101 differentially expressed metabolites, including 62 upregulated and 39 downregulated metabolites. KEGG pathway enrichment analysis of both proteomic and metabolomic data revealed seven key signalling pathways, 11 key proteins, and 26 key metabolites that interact through metabolic and organic system networks. Next, we examined the disease progression of E. granulosus infection in GHSR-knockout mice. Compared with the E. granulosus (Eg) group, the GHSR-KO group presented a significant reduction in the number of liver infection foci. The serum and liver ghrelin levels were significantly greater in the E. granulosus group than in the control group, along with increased secretion of proinflammatory cytokines (IL-2 and IFN-γ) and decreased secretion of anti-inflammatory cytokines (IL-4 and IL-10). In contrast, the GHSR-KO group presented significantly lower ghrelin levels in both the serum and liver, with reduced proinflammatory cytokine secretion and increased anti-inflammatory cytokine secretion, similar to those of the control group. Furthermore, ghrelin and inflammation-related factors, including MyD88, NF-κB p65, iNOS, and Arg-1, exhibited coordinated expression changes in liver lesions and surrounding areas. These findings suggest that GHSR gene knockout can ameliorate the progression of liver E. granulosus infection and associated liver inflammation.
Collapse
Affiliation(s)
- Jiang Zhu
- Department of Abdominal Surgery, The Third People Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- School of Medicine, Shihezi University, Shihezi, 832003, China
| | - Tanfang Zhou
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Guangfeng Chen
- Department of Abdominal Surgery, The Third People Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- School of Medicine, Shihezi University, Shihezi, 832003, China
| | - Huijing Gao
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Xia Chen
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Ayinula Tuohetali
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Ya Song
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Dongming Pang
- Department of Abdominal Surgery, The Third People Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China.
| | - Kalibixiati Aimulajiang
- State Κey Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
32
|
Cong M, Li S, Fu Y, Wang T, Wei H, Ma X, Liu F, Sheng X, Long K, Hou H, Sun X, Li W, Zhang L. Ghrelin increases cis-platinum resistance and promotes aggressiveness of osteosarcoma by activating AKT and Wnt/β-catenin pathways. J Orthop Surg Res 2025; 20:253. [PMID: 40059184 PMCID: PMC11892314 DOI: 10.1186/s13018-024-05261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/11/2024] [Indexed: 05/13/2025] Open
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy because of its extra high tendency of metastasis. In-depth research is needed to uncover the pathogenesis of patients with OS cells. We collected 74 tissue samples from patients with OS cells and measured the expression levels of ghrelin by immunohistochemistry. Ghrelin was added into OS cell lines in CCK8 assays, JC-1 staining and Western blot analysis were performed to explore its effect on the aggressiveness of OS cells and drug resistance. To determine its function, ghrelin was overexpressed or knocked down in OS cells and then detect cell proliferation in the xenograft mouse model and orthotopic model. Western blot analysis was performed to explore ghrelin-regulated signal pathways. In this work, we identified the relation between the level of ghrelin expression and poor prognosis of OS patients. As well as promoting proliferation, migration, and invation, ghrelin promotes the survival of OS in vitro as well as in vivo, and reduces the apoptosis of OS cells. What's more, ghrelin increases the resistance of cis-platinum by changing mitochondrial function and decreases the expression of MDR-1. Above all, these results demonstrated ghrelin exerts tumorigenic and metastatic effects and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Menglin Cong
- Department of Spine Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Shufeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yu Fu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Ting Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Huizhen Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaojie Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fangming Liu
- Department of Rheumatology and immunology, Shandong Provincial Qianfoshan Hospital, Institute of Anesthesia and Respiratory Critical Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - XieHuang Sheng
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, China
| | - Kehan Long
- Clinical Medical College of Weifang Medical University, Weifang, China
| | - Haocheng Hou
- College of Basic Medicine, Shandong University, Jinan, China
| | - Xuecheng Sun
- Department of Orthopedic Trauma, Weifang People's Hospital, Weifang, China.
| | - Weiwei Li
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China.
| | - Lei Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
- Tissue Engineering Laboratory, Department of Radiology, Shandong First Medical University, Jinan, China.
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China.
| |
Collapse
|
33
|
Nomura K, Yamasaki Y, Takeji K, Deha S, Yamashita K, Izumi-Mishima Y, Yasui-Yamada S, Kuroda M, Harada N, Kitamura T, Tsutsumi YM, Tsutsumi R, Nakaya Y, Sakaue H. Gut-pancreas axis in the control of insulin secretion in streptozotocin-resistant rats. Biochem Biophys Res Commun 2025; 752:151487. [PMID: 39955952 DOI: 10.1016/j.bbrc.2025.151487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
The Spontaneously Running Tokushima Shikoku (SPORTS) rat is characterized by hyperactive behavior but is also a potential model for studies of the role of the gut-pancreas axis in the regulation of insulin secretion. We here explored the role of ghrelin, a hormone associated with appetite regulation, in insulin dynamics within the context of streptozotocin (STZ) resistance in SPORTS rats. Comprehensive analyses-including histological examinations, gene expression profiling, and assessment of metabolic parameters-revealed that SPORTS rats are resistant to STZ-induced pancreatic injury and that, in addition to low circulating ghrelin levels, they manifest increased circulating levels of active glucagon-like peptide-1 (GLP-1) and upregulated expression of Pdx1 in the pancreas. Ghrelin administration attenuated the STZ resistance of these rats, with suppression of GLP-1 and downregulation of Pdx1 expression being implicated in this effect. Our results highlight the complex interplay among ghrelin, GLP-1, and insulin dynamics, and they suggest potential new therapeutic targets for diabetes.
Collapse
Affiliation(s)
- Kazuhiro Nomura
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Yamasaki
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Fukuoka Women's University, Fukuoka, Japan
| | - Kana Takeji
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Sachie Deha
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kaho Yamashita
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuna Izumi-Mishima
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Sonoko Yasui-Yamada
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Nagakatsu Harada
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Yasuo M Tsutsumi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Yutaka Nakaya
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Diabetes Therapeutics and Research Center, University of Tokushima, Tokushima, Japan.
| |
Collapse
|
34
|
Tatsuta R, Tanaka R, Tashibu A, Suzuki Y, Suzuki K, Shibata T, Ando T, Shin T, Sato Y, Itoh H. Association of chemotherapy-induced nausea and vomiting or anorexia with plasma levels of five gastrointestinal peptides in patients receiving chemotherapy. J Pharm Health Care Sci 2025; 11:17. [PMID: 40045433 PMCID: PMC11881272 DOI: 10.1186/s40780-025-00424-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Imbalance between gastrointestinal peptides has been implicated as a cause of chemotherapy-induced nausea and vomiting (CINV) and anorexia in cancer patients. This study comprehensively evaluated the changes in blood levels of five gastrointestinal peptide: substance P, neuropeptide (NPY), motilin, ghrelin and leptin, following chemotherapy, and the relationship between these peptides and CINV or anorexia. METHODS This single-center, prospective, observational study recruited 20 patients with esophageal cancer, urothelial cancer, or testiculoma undergoing cisplatin-based chemotherapy. Plasma levels of five gastrointestinal peptides were measured on days 1 (baseline; before administering chemotherapy), 3, 5 and 8 of the chemotherapy session. Anorexia and CINV were defined as visual analog scale scores 25 mm or higher at least once during the observation period. RESULTS Plasma NPY and leptin were significantly elevated in the early phase (day 3) of the chemotherapy session, while plasma motilin and substance P were significantly elevated in the late phase (days 5 and 8). Plasma motilin showed significant elevation on days 5 and 8 compared to baseline in CINV group but no significant increase in non-CINV group, and the levels were significantly higher in CINV than in non-CINV group. Plasma leptin peaked significantly on day 3 in both anorexia and non-anorexia groups, and remained significantly higher on day 5 compared to baseline in anorexia group but not in non-anorexia group. CONCLUSION CINV is associated with excessive secretion of motilin and anorexia is related to sustained elevation of leptin, suggesting the potential of these peptides as quantitative indicators of CINV and anorexia.
Collapse
Affiliation(s)
- Ryosuke Tatsuta
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan.
| | - Ryota Tanaka
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Asami Tashibu
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Yosuke Suzuki
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Kosuke Suzuki
- Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Tomotaka Shibata
- Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Tadasuke Ando
- Department of Urology, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Toshitaka Shin
- Department of Urology, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Yuhki Sato
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Hiroki Itoh
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| |
Collapse
|
35
|
Shiimura Y, Im D, Tany R, Asada H, Kise R, Kurumiya E, Wakasugi-Masuho H, Yasuda S, Matsui K, Kishikawa JI, Kato T, Murata T, Kojima M, Iwata S, Masuho I. The structure and function of the ghrelin receptor coding for drug actions. Nat Struct Mol Biol 2025; 32:531-542. [PMID: 39833471 DOI: 10.1038/s41594-024-01481-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Drugs targeting the ghrelin receptor hold therapeutic potential in anorexia, obesity and diabetes. However, developing effective drugs is challenging. To tackle this common issue across a broad drug target, this study aims to understand how anamorelin, the only approved drug targeting the ghrelin receptor, operates compared to other synthetic drugs. Our research elucidated the receptor's structure with anamorelin and miniGq, unveiling anamorelin's superagonistic activity. We demonstrated that ligands with distinct chemical structures uniquely bind to the receptor, resulting in diverse conformations and biasing signal transduction. Moreover, our study showcased the utility of structural information in effectively identifying natural genetic variations altering drug action and causing severe functional deficiencies, offering a basis for selecting the right medication on the basis of the individual's genomic sequence. Thus, by building on structural analysis, this study enhances the foundational framework for selecting therapeutic agents targeting the ghrelin receptor, by effectively leveraging signaling bias and genetic variations.
Collapse
Affiliation(s)
- Yuki Shiimura
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan.
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Dohyun Im
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Tany
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Hidetsugu Asada
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryoji Kise
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Eon Kurumiya
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | | | - Satoshi Yasuda
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
- Membrane Protein Research Center, Chiba University, Chiba, Japan
| | - Kazuma Matsui
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Jun-Ichi Kishikawa
- Institute for Protein Research, Osaka University, Osaka, Japan
- Faculty of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | - Takayuki Kato
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
- Membrane Protein Research Center, Chiba University, Chiba, Japan
| | - Masayasu Kojima
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Ikuo Masuho
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA.
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA.
| |
Collapse
|
36
|
Kaiya H, Nobata S, Takei Y. Ghrelin suppresses water intake with a different physiological significance from atrial natriuretic peptide in conscious seawater-acclimated eels. J Exp Biol 2025; 228:JEB249707. [PMID: 39882695 DOI: 10.1242/jeb.249707] [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: 10/16/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
In general, ghrelin is known as one of the orexigenic (increasing appetite or food intake) hormones in mammals. However, it has also been shown that ghrelin inhibits water intake, which appears to be inconsistent with its role in the feeding response. In this study, the effect of ghrelin on water intake was comprehensively addressed using conscious seawater-acclimated eels as an experimental model for water drinking behaviour. When injected intra-arterially, ghrelin inhibited copious drinking in a dose-dependent manner without affecting arterial pressure. This effect contrasted with the inhibitory effect of atrial natriuretic peptide (ANP) on drinking, which is synchronized with a vasodepressor effect. Similarly, intra-cerebroventricular injection of ghrelin also decreased the drinking rate without affecting arterial pressure. Continuous infusion of ghrelin from the ventral aorta also decreased the drinking rate, concomitant with an increase in plasma ghrelin concentration. The inhibitory effects of ghrelin on drinking were as potent and efficacious as those of ANP. The inhibitory action was not blocked by pre-treatment with a ghrelin receptor antagonist ([D-Lys3] GHRP-6); consistently, the agonist form (GHRP-6) injected intra-arterially did not show any inhibitory effect of ghrelin when injected peripherally. These results demonstrate that ghrelin is a potent anti-dipsogen in eels without baroreflex and ANP secretion, and it is possible that ghrelin's effect might be mediated through another type of ghrelin receptor that [D-Lys3] GHRP-6 or GHRP-6 do not bind.
Collapse
Affiliation(s)
- Hiroyuki Kaiya
- Faculty of Science, University of Toyama, 3190 Gofuku, Toyama-city, Toyama 930-8555, Japan
- Department of Biochemistry, National Cardiovascular Centre Research Institute, 6-1 Kishibe-Shinmachi, Suita-city 564-8565, Japan
| | - Shigenori Nobata
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa, Chiba 277-8564, Japan
| | - Yoshio Takei
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa, Chiba 277-8564, Japan
| |
Collapse
|
37
|
Prost S, Elbers JP, Slezacek J, Hykollari A, Fuselli S, Smith S, Fusani L. The unexpected loss of the 'hunger hormone' ghrelin in true passerines: a game changer in migration physiology. ROYAL SOCIETY OPEN SCIENCE 2025; 12:242107. [PMID: 40109942 PMCID: PMC11919490 DOI: 10.1098/rsos.242107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Migratory birds must accumulate large amounts of fat prior to migration to sustain long flights. In passerines, the small body size limits the amount of energy stores that can be transported, and therefore birds undergo cycles of extreme fattening and rapid exhaustion of reserves. Research on these physiological adaptations was rattled by the discovery that birds have lost the main vertebrate regulator of fat deposition, leptin. Recent studies have thus focused on ghrelin, known as 'hunger hormone', a peptide secreted by the gastrointestinal tract to regulate, e.g. food intake and body mass in vertebrates. Studies on domestic species showed that, in birds, ghrelin has effects opposite to those described in mammals such as inhibiting instead of promoting food intake. Furthermore, recent studies have shown that ghrelin administration influences migratory behaviour in passerine birds. Using comparative genomics and immunoaffinity chromatography, we show that ghrelin has been lost in Eupasseres after the basic split from Acanthisitti about 50 Ma. We found that the ghrelin receptor is still conserved in passerines. The maintenance of a functional receptor system suggests that in Eupasserines, another ligand has replaced ghrelin, perhaps to bypass the feedback system that would hinder the large pre-migratory accumulation of subcutaneous fat.
Collapse
Affiliation(s)
- Stefan Prost
- Ecology and Genetics Research Unit, University of Oulu, Oulu, Finland
| | - Jean P Elbers
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Julia Slezacek
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Alba Hykollari
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Silvia Fuselli
- Life Sciences and Biotechnologies, University of Ferrara, Ferrara, Italy
| | - Steve Smith
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Leonida Fusani
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- Department of Behavioral and Cognitive Biology, University of Vienna, Vienna, Austria
| |
Collapse
|
38
|
Xin C, Yao J, Li H, Sun X, Wang H. Relationship between ghrelin and thyroid disease: a meta-analysis. Front Endocrinol (Lausanne) 2025; 16:1505085. [PMID: 40093748 PMCID: PMC11906317 DOI: 10.3389/fendo.2025.1505085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Background Previous studies have identified a close relationship between ghrelin levels and thyroid disease. Ghrelin levels were lower in patients with hyperthyroidism compared with healthy individuals, and increased after treatment. However, other studies have reported inconsistent results. As such, the association between ghrelin and thyroid disease remains controversial. Methods A literature search of the Web of Science, Wiley Online Library, Embase, and PubMed databases was performed. The title or abstract search term "thyroid" was used in combination with "ghrelin". Meta-analysis results are reported as standardized mean difference with corresponding 95% confidence interval (CI). Results Twenty-three studies were included in this meta-analysis. Ghrelin levels in patients with hyperthyroidism were significantly lower than those in healthy individuals (SMD: -1.03, 95% CI [-1.75, 0.32]), but significantly higher after effective treatment (SMD: 0.77, 95% CI [0.03, 1.51]). Ghrelin levels were higher, but not significantly, in patients with hypothyroidism compared with healthy controls (SMD: 0.48, 95% CI [-0.13, 1.08]). Conclusions This systematic review is the first to evaluate the relationship between ghrelin and thyroid disease. Determining the role of ghrelin in thyroid disease will significantly contribute to understand of symptom or pathomechanism. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024591501.
Collapse
Affiliation(s)
- Caihong Xin
- Department of Endocrinology and Metabolism, Fourth People’s Hospital of Shenyang, Shenyang, China
| | - Jiayi Yao
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huijuan Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Sun
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huijuan Wang
- Department of Diabetes, Taiping Street Health Center of Xiangcheng District of Suzhou City, Suzhou, China
| |
Collapse
|
39
|
Edwards A, DeSante S, Spencer CD, Hyland L, Smith A, Sankhe AS, Szilvásy-Szabó A, Fekete C, Hill MN, Chee MJ, Abizaid A. Ghrelin Recruits the Endocannabinoid System to Modulate Food Reward. J Neurosci 2025; 45:e1620242024. [PMID: 39779372 PMCID: PMC11867019 DOI: 10.1523/jneurosci.1620-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/05/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Ghrelin enhances feeding by activating the growth hormone secretagogue receptor (GHSR). In the brain, GHSRs are expressed in regions responsible for regulating food motivation including the ventral tegmental area (VTA). Endogenous cannabinoids also promote food-seeking behaviors through the cannabinoid receptor-1 (CB-1Rs) in brain regions including the VTA. It is not known, however, if ghrelin and endocannabinoids interact in the VTA to produce these effects. We therefore examined if GHSR and CB-1R interact within the VTA to enhance food motivation. Results show that GHSR and CB-1R mRNA are expressed in the VTA cells in male and female rats and mice, with the GHSR being expressed in dopamine cells and the CB-1R being expressed primarily in nondopaminergic cells with no obvious sex differences. Ghrelin directly activated and increased excitatory tone onto dopamine cells of male and female mice. Male rats lacking fully functional GHSR signaling showed disrupted gene expression of transcripts important for regulating the synthesis, release, and degradation of endocannabinoids and lowered the levels of 2-arachidonoylglycerol (2-AG) within the VTA. Moreover, pharmacological antagonism of VTA CB-1Rs attenuates the orexigenic and appetitive effects of intra-VTA ghrelin in rats and blocks the ability of ghrelin to promote excitatory drive to VTA dopamine neurons. Finally, blocking the breakdown of cannabinoids in the VTA enhances the effects of ghrelin on food motivation. Together, our data show that ghrelin stimulates VTA dopamine cells and ultimately food motivation in part through a mechanism that involves endocannabinoid signaling at the CB-1R.
Collapse
Affiliation(s)
- Alexander Edwards
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Stephanie DeSante
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Carl Duncan Spencer
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Lindsay Hyland
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Andrea Smith
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Aditi S Sankhe
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Anett Szilvásy-Szabó
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest H-1083, Hungary
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest H-1083, Hungary
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N4T1, Canada
| | - Melissa J Chee
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Alfonso Abizaid
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
40
|
Mengr A, Šmotková Z, Pačesová A, Železná B, Kuneš J, Maletínská L. Reduction of Neuroinflammation as a Common Mechanism of Action of Anorexigenic and Orexigenic Peptide Analogues in the Triple Transgenic Mouse Model of Alzheimer´s Disease. J Neuroimmune Pharmacol 2025; 20:18. [PMID: 39932627 PMCID: PMC11813825 DOI: 10.1007/s11481-025-10174-w] [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: 10/13/2024] [Accepted: 01/24/2025] [Indexed: 02/14/2025]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Characterized by progressive neurodegeneration, AD typically begins with mild cognitive decline escalating to severe impairment in communication and responsiveness. It primarily affects cerebral regions responsible for cognition, memory, and language processing, significantly impeding the functional independence of patients. With nearly 50 million dementia cases worldwide, a number expected to triple by 2050, the need for effective treatments is more urgent than ever. Recent insights into the association between obesity, type 2 diabetes mellitus, and neurodegenerative disorders have led to the development of promising treatments involving antidiabetic and anti-obesity agents. One such novel promising candidate for addressing AD pathology is a lipidized analogue of anorexigenic peptide called prolactin-releasing peptide (palm11-PrRP31). Interestingly, anorexigenic and orexigenic peptides have opposite effects on food intake regulation, however, both types exhibit neuroprotective properties. Recent studies have also identified ghrelin, an orexigenic peptide, as a potential neuroprotective agent. Hence, we employed both anorexigenic and orexigenic compounds to investigate the common mechanisms underpinning their neuroprotective effects in a triple transgenic mouse model of AD (3xTg-AD mouse model) combining amyloid-beta (Aβ) pathology and Tau pathology, two hallmarks of AD. We treated 3xTg-AD mice for 4 months with two stable lipidized anorexigenic peptide analogues - palm11-PrRP31, and liraglutide, a glucagon-like peptide 1 (GLP-1) analogue - as well as Dpr3-ghrelin, a stable analogue of the orexigenic peptide ghrelin, and using the method of immunohistochemistry and western blot demonstrate the effects of these compounds on the development of AD-like pathology in the brain. Palm11-PrRP31, Dpr3-ghrelin, and liraglutide reduced intraneuronal deposits of Aβ plaque load in the hippocampi and amygdalae of 3xTg-AD mice. Palm11-PrRP31 and Dpr3-ghrelin reduced microgliosis in the hippocampi, amygdalae, and cortices of 3xTg-AD mice. Palm11-PrRP31 and liraglutide reduced astrocytosis in the amygdalae of 3xTg-AD mice. We propose that these peptides are involved in reducing inflammation, a common mechanism underlying their therapeutic effects. This is the first study to demonstrate improvements in AD pathology following the administration of both orexigenic and anorexigenic compounds, highlighting the therapeutic potential of food intake-regulating peptides in neurodegenerative disorders.
Collapse
Affiliation(s)
- Anna Mengr
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Zuzana Šmotková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Kateřinská 32, 12108, Prague, Czech Republic
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00, Prague, Czech Republic
| | - Andrea Pačesová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00, Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic.
| |
Collapse
|
41
|
Ma Y, Zhang H, Yan Q, Wang P, Guo W, Yu L. The antidiabetic effect of safflower yellow by regulating the GOAT/ghrelin/GHS-R1a/cAMP/TRPM2 pathway. Sci Rep 2025; 15:5037. [PMID: 39934157 PMCID: PMC11814266 DOI: 10.1038/s41598-025-87201-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
Safflower yellow (SY), derived from Carthamus tinctorius L., is a valuable natural edible pigment that exhibits anti-type 2 diabetes mellitus (T2DM) efficacy; however, its mechanism of action is unclear, which hinders its effective use. In this study, we examined the impact of SY on glucose metabolism and insulin secretion both in vivo and in vitro and elucidated the possible underlying mechanism. First, molecular docking demonstrated a strong binding affinity between SY and ghrelin O-acyltransferase (GOAT) protein, which was validated by a cell heat transfer assay (CETSA) and drug affinity response target stability (DARTS) in MIN6 cells. In MIN6 cells, SY increased insulin secretion and showed time- and dose-dependent inhibition of GOAT expression and acyl ghrelin (AG) secretion without affecting the overall levels of ghrelin. Furthermore, ELISA revealed that SY enhanced high glucose (HG)-induced insulin secretion, and immunofluorescence revealed the co-localization of GOAT and ghrelin in MIN6 cells, which was suppressed by SY treatment. The mechanism analysis by Western blot demonstrated that SY downregulated the protein levels of GOAT and GHS-R1a in MIN6 cells while increasing HG-stimulated cAMP and activation of transient receptor potential melastatin 2 (TRPM2). In in vivo experiments, the intraperitoneal injection of SY significantly improved pathological damage to the pancreas, glucose tolerance, and insulin resistance in a mouse model of high-fat diet (HFD)/streptozotocin (STZ)-induced T2DM in a dose-dependent manner. SY enhanced insulin secretion by inhibiting the GOAT/ghrelin system in vivo. In conclusion, we demonstrated that SY exhibits an observable protective effect on diabetes through the GOAT/ghrelin/GHS-R1a/cAMP/TRPM2 pathway. Our findings provide a basis for further investigation of the hypoglycemic mechanism of SY and its potential for further development and utilization.
Collapse
Affiliation(s)
- Yunxiao Ma
- Department of Endocrinology, Interventional Therapy, and Otolaryngology-Head and Neck Surgery of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Haifeng Zhang
- Department of Endocrinology, Interventional Therapy, and Otolaryngology-Head and Neck Surgery of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Qihui Yan
- Department of Endocrinology, Interventional Therapy, and Otolaryngology-Head and Neck Surgery of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Ping Wang
- Department of Endocrinology, Interventional Therapy, and Otolaryngology-Head and Neck Surgery of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Weiying Guo
- Department of Endocrinology, Interventional Therapy, and Otolaryngology-Head and Neck Surgery of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130000, China.
| | - Lu Yu
- Department of Endocrinology, Interventional Therapy, and Otolaryngology-Head and Neck Surgery of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130000, China.
| |
Collapse
|
42
|
Tiezzi M, Vieceli Dalla Sega F, Gentileschi P, Campanelli M, Benavoli D, Tremoli E. Effects of Weight Loss on Endothelium and Vascular Homeostasis: Impact on Cardiovascular Risk. Biomedicines 2025; 13:381. [PMID: 40002792 PMCID: PMC11853214 DOI: 10.3390/biomedicines13020381] [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: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Available knowledge shows that obesity is associated with an impaired endothelial function and an increase in cardiovascular risk, but the mechanisms of this association are not yet fully understood. Adipose tissue dysfunction, adipocytokines production, along with systemic inflammation and associated comorbidities (e.g., diabetes and hypertension), are regarded as the primary physiological and pathological factors. Various strategies are now available for the control of excess body weight. Dietary regimens alone, or in association with bariatric surgery when indicated, are now widely used. Of particular interest is the understanding of the effect of these interventions on endothelial homeostasis in relation to cardiovascular health. Substantial weight loss resulting from both diet and bariatric surgery decreases circulating biomarkers and improves endothelial function. Extensive clinical trials and meta-analyses show that bariatric surgery (particularly gastric bypass) has more substantial and long-lasting effect on weight loss and glucose regulation, as well as on distinct circulating biomarkers of cardiovascular risk. This review summarizes the current understanding of the distinct effects of diet-induced and surgery-induced weight loss on endothelial function, focusing on the key mechanisms involved in these effects.
Collapse
Affiliation(s)
- Margherita Tiezzi
- Dipartimento Cardiovascolare, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy;
| | | | - Paolo Gentileschi
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
- Dipartimento di Scienze Chirurgiche, Università di Roma Tor Vergata, 00133 Roma, Italy
| | - Michela Campanelli
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
| | - Domenico Benavoli
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
| | - Elena Tremoli
- Dipartimento Cardiovascolare, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy;
| |
Collapse
|
43
|
Poelman R, Le May MV, Schéle E, Stoltenborg I, Dickson SL. Intranasal Delivery of a Ghrelin Mimetic Engages the Brain Ghrelin Signaling System in Mice. Endocrinology 2025; 166:bqae166. [PMID: 39813130 PMCID: PMC11795113 DOI: 10.1210/endocr/bqae166] [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: 09/26/2024] [Revised: 11/27/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
Ghrelin, the endogenous ligand of the growth hormone secretagogue receptor (GHSR), promotes food intake and other feeding behaviors, and stimulates growth hormone (GH) release from the pituitary. Growth hormone secretagogues (GHS), such as GHRP-6 and MK-0677, are synthetic GHSR ligands that activate orexigenic neuropeptide Y neurons that coexpress agouti-related peptide (AgRP) in the arcuate nucleus of the hypothalamus when administered systemically. Systemic GHRP-6 also stimulates GH release in humans and rats. Thus, GHS and ghrelin have therapeutic relevance in patients who could benefit from its orexigenic and/or GH-releasing effects. This study examined whether intranasal delivery of ghrelin, GHRP-6, or MK-0677 engages the brain ghrelin signaling system. Effective compounds and doses were selected based on increased food intake after intranasal application in mice. Only GHRP-6 (5 mg/kg) increased food intake without adverse effects, prompting detailed analysis of meal patterns, neuronal activation in the arcuate nucleus (via Fos mapping) and neurochemical identification of c-fos messenger RNA (mRNA)-expressing neurons using RNAscope. We also assessed the effect of intranasal GHRP-6 on serum GH levels. Intranasal GHRP-6 increased food intake by increasing meal frequency and size. Fos expression in the arcuate nucleus was higher in GHRP-6-treated mice than in saline controls. When examining the neurochemical identity of c-fos-mRNA-expressing neurons, we found coexpression with 63.5 ± 1.9% Ghsr mRNA, 79 ± 6.8% Agrp mRNA, and 11.4 ± 2.5% Ghrh mRNA, demonstrating GHRP-6's ability to engage arcuate nucleus neurons involved in food intake and GH release. Additionally, intranasal GHRP-6 elevated GH serum levels. These findings suggest that intranasal GHRP-6, but not ghrelin or MK-0677, can engage the brain ghrelin signaling system.
Collapse
Affiliation(s)
- Renée Poelman
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-413 90 Gothenburg, Sweden
| | - Marie V Le May
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-413 90 Gothenburg, Sweden
| | - Erik Schéle
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-413 90 Gothenburg, Sweden
| | - Iris Stoltenborg
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-413 90 Gothenburg, Sweden
| | - Suzanne L Dickson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-413 90 Gothenburg, Sweden
| |
Collapse
|
44
|
Ford AL, Taft CW, Sprague-Getsy AM, Carlson GC, Mate NA, Sieburg MA, Chisholm JD, Hougland JL. A Modular Customizable Ligand-Conjugate (LC) System Targeting Ghrelin O-Acyltransferase. Biomolecules 2025; 15:204. [PMID: 40001510 PMCID: PMC11852496 DOI: 10.3390/biom15020204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 02/27/2025] Open
Abstract
Ghrelin is a 28 amino acid peptide hormone that impacts a wide range of biological processes, including appetite regulation, glucose metabolism, growth hormone regulation, and cognitive function. To bind and activate its cognate receptor, ghrelin must be acylated on a serine residue in a post-translational modification performed by ghrelin O-acyltransferase (GOAT). GOAT is a membrane-bound O-acyltransferase (MBOAT) responsible for the catalysis of the addition of an octanoyl fatty acid to the third serine of desacyl ghrelin. Beyond its canonical role for ghrelin maturation in endocrine cells within the stomach, GOAT was recently reported to be overexpressed in prostate cancer (PCa) cells and detected at increased levels in the serum and urine of PCa patients. This suggests GOAT can serve as a potential route for the detection and therapeutic targeting of PCa and other diseases that exhibit GOAT overexpression. Building upon a ghrelin mimetic peptide with nanomolar affinity for GOAT, we developed an antibody-conjugate-inspired system for customizable ligand-conjugate (LC) synthesis allowing for the attachment of a wide range of cargoes. The developed synthetic scheme allows for the easy synthesis of the desired LCs and demonstrates that our ligand system tolerates an extensive palette of cargoes while maintaining nanomolar affinity against GOAT.
Collapse
Affiliation(s)
- Amber L. Ford
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Caine W. Taft
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Andrea M. Sprague-Getsy
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Gracie C. Carlson
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Nilamber A. Mate
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - Michelle A. Sieburg
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
| | - John D. Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
- BioInspired Syracuse, Syracuse University, Syracuse, NY 13244, USA
| | - James L. Hougland
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; (A.L.F.); (C.W.T.); (A.M.S.-G.); (G.C.C.); (N.A.M.); (M.A.S.); (J.D.C.)
- BioInspired Syracuse, Syracuse University, Syracuse, NY 13244, USA
- Department of Biology, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
45
|
Alogaiel DM, Alsuwaylihi A, Alotaibi MS, Macdonald IA, Lobo DN. Effects of Ramadan intermittent fasting on hormones regulating appetite in healthy individuals: A systematic review and meta-analysis. Clin Nutr 2025; 45:250-261. [PMID: 39842253 DOI: 10.1016/j.clnu.2025.01.005] [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: 11/11/2024] [Revised: 12/17/2024] [Accepted: 01/05/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND AND AIMS This systematic review and meta-analysis aimed to examine the effect of Ramadan intermittent fasting on appetite-regulating hormones including leptin, ghrelin, insulin, gastrin, glucagon-like peptide-1, peptide YY, and cholecystokinin. METHODS We searched the MEDLINE, Embase, Cochrane Library, CINAHL, Google Scholar, and Web of Science databases to identify relevant research on appetite-regulating hormones during Ramadan intermittent fasting, published until the end of March 2024. RESULTS Data from 16 eligible studies comprising 664 participants (341, 51.4 % male) with a mean ± standard deviation age of 33.9 ± 10.8 years were included. The meta-analysis included 12 studies with complete leptin data, showing no significant effect of Ramadan intermittent fasting on leptin concentrations (standardised mean difference - SMD = -0.11 μg/mL, 95 % CI: -0.36 to 0.14). Analysis of three studies with complete ghrelin data demonstrated a significant increase in ghrelin concentrations following Ramadan intermittent fasting (SMD = 0.31 pg/mL, 95 % CI: 0.03 to 0.60). Six studies examining insulin concentrations pre- and post-fasting revealed no significant effect on insulin concentrations (SMD = -0.24 μU/mL, 95 % CI: -0.54 to 0.02). Similarly, analysis of three studies with complete gastrin data showed no significant effect of intermittent fasting on gastrin concentrations (SMD = 0.23 pg/mL, 95 % CI: -0.71 to 0.99). CONCLUSION Ramadan intermittent fasting significantly increases ghrelin concentrations while showing no significant effects on leptin, insulin, and gastrin. While ghrelin findings were consistent across studies, the high heterogeneity in leptin studies suggests further research to better understand the effects of Ramadan intermittent fasting on appetite-regulating hormones.
Collapse
Affiliation(s)
- Deema M Alogaiel
- Nottingham Digestive Diseases Centre, Division of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK; Health Sciences Department, College of Health and Rehabilitation, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Abdulaziz Alsuwaylihi
- Nottingham Digestive Diseases Centre, Division of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; Department of Clinical Nutrition, King Saud Medical City, Ministry of Health, Riyadh, Saudi Arabia
| | - May S Alotaibi
- Health Sciences Department, College of Health and Rehabilitation, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Division of Food, Nutrition & Dietetics, School of Biosciences, University of Nottingham, LE12 5RD, UK
| | - Ian A Macdonald
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Dileep N Lobo
- Nottingham Digestive Diseases Centre, Division of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK; Division of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Sixten HJ, Rønnestad I, Bogevik AS, Aspevik T, Oterhals Å, Gomes AS, Lai F, Tolås I, Gelebard V, Hillestad M, Kousoulaki K. Side-Stream Based Marine Solubles From Atlantic Cod ( Gadus morhua) Modulate Appetite and Dietary Nutrient Utilization in Atlantic Salmon ( Salmo salar L.) and can Replace Fish Meal. AQUACULTURE NUTRITION 2025; 2025:4872889. [PMID: 39949357 PMCID: PMC11824393 DOI: 10.1155/anu/4872889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025]
Abstract
Whitefish fisheries' side-stream biomass is an abundant underutilized resource that can be valorized to benefit future aquaculture sustainability. Four novel ingredients based on side-streams from Atlantic cod (Gadus morhua) fileting were produced. FM-hb, a fish meal (FM), and FPH-hb, a fish protein hydrolysate based on heads (h) and backbones (b); FM-hbg, a FM based on heads, backbones, and viscera/guts (g); and FPC-g, a fish protein concentrate based on viscera preserved in formic acid. Four diets were prepared containing one of the ingredients replacing 50% of the dietary FM protein, in addition to a positive (FM10) and a negative (FM5) control. The six diets were fed to triplicate tanks with Atlantic salmon (Salmo salar L.; 113 ± 1 g) over 8 weeks. Besides general performance, gut and brain gene expression for selected hormones and key neuropeptides involved in the control of appetite and digestive processes were studied during feeding and postprandial, and possible reference levels for Atlantic salmon were established. All side-stream-added diets performed well, with no significant differences in performance and biometrics between the treatments. Some gene expression differences were observed, but no well-defined patterns emerged supporting clear dietary effects related to digestive performance or appetite. However, in the brain, a short-time upregulation of agouti-related protein-1 (agrp1), corresponded to higher cumulative feed intake (FI) for the FM10 diet supporting notions that this may be a candidate biomarker for appetite in salmon. Expression of stomach ghrelin-1 (ghrl1) was higher than ghrelin-2 (ghrl2) and membrane-bound O-acyltransferase domain-containing 4 (mboat4), and midgut peptide YYa-2 (pyya2) and glucagon-a (gcga) were higher than peptide YYb-1 (pyyb1). A comparison showed that midgut peptide YYa-1 (pyya1), pyya2, and gcga expressions were higher than in the hindgut, which is opposite of what is found in mammals. In conclusion, this study shows that sustainable side-stream raw materials with different characteristics can partly replace high-quality commercial FMs giving similar performance.
Collapse
Affiliation(s)
- Hanne Jorun Sixten
- Department of Research and Development, BioMar AS, Trondheim, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Ivar Rønnestad
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - André S. Bogevik
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Tone Aspevik
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Åge Oterhals
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Ana S. Gomes
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Institute of Marine Research, Tromsø, Norway
| | - Floriana Lai
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Ingvill Tolås
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Department of Biological Sciences, NTNU Ålesund, Ålesund, Norway
| | - Virginie Gelebard
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Marie Hillestad
- Department of Research and Development, BioMar AS, Trondheim, Norway
| | | |
Collapse
|
47
|
Wang HJ, Fan W, Liu S, Kim K, Matsushima A, Ogawa S, Kang HG, Zhu J, Estepa G, He M, Crossley L, Liddle C, Kim MS, Truitt ML, Yu RT, Atkins AR, Downes M, Evans RM. BCL6 coordinates muscle mass homeostasis with nutritional states. Proc Natl Acad Sci U S A 2025; 122:e2408896122. [PMID: 39841144 PMCID: PMC11789089 DOI: 10.1073/pnas.2408896122] [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: 05/06/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Nutritional status is a determining factor for growth during development and homeostatic maintenance in adulthood. In the context of muscle, growth hormone (GH) coordinates growth with nutritional status; however, the detailed mechanisms remain to be fully elucidated. Here, we show that the transcriptional repressor B cell lymphoma 6 (BCL6) maintains muscle mass by sustaining GH action. Muscle-specific genetic deletion of BCL6 at either perinatal or adult stages profoundly reduces muscle mass and compromises muscle strength. Conversely, muscle-directed viral overexpression of BCL6 significantly reverses the loss of muscle mass and strength. Mechanistically, we show that BCL6 transcriptionally represses the suppressor of cytokine signaling 2 to sustain the anabolic actions of GH in muscle. Additionally, we find that GH itself transcriptionally inhibits BCL6 through the Janus kinase and signal transducer and activator of transcription 5 (JAK/STAT5) pathway. Supporting the physiologic relevance of this feedback regulation, we show the coordinated suppression of muscle Bcl6 expression with the induction of GH in the fasted state. These findings reveal the complexity of the feedback controls modulating GH signaling and identify BCL6 as a key homeostatic regulator coordinating muscle mass with nutrient availability. Moreover, these studies open avenues for targeted therapeutic strategies to combat muscle-wasting conditions.
Collapse
Affiliation(s)
- Hui J. Wang
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Weiwei Fan
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Sihao Liu
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Kyeongkyu Kim
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Ayami Matsushima
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka819-0395, Japan
| | - Satoshi Ogawa
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Hyun Gyu Kang
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Jonathan Zhu
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Gabriela Estepa
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Mingxiao He
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Lillian Crossley
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney Medical School, University of Sydney, Westmead, NSW2124, Australia
| | - Minseok S. Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Morgan L. Truitt
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | | | - Michael Downes
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute, La Jolla, CA92037-1002
| |
Collapse
|
48
|
Chen K, Wu X, Li X, Pan H, Zhang W, Shang J, Di Y, Liu R, Zheng Z, Hou X. Antimicrobial Neuropeptides and Their Receptors: Immunoregulator and Therapeutic Targets for Immune Disorders. Molecules 2025; 30:568. [PMID: 39942672 PMCID: PMC11820534 DOI: 10.3390/molecules30030568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
The interaction between the neuroendocrine system and the immune system plays a key role in the onset and progression of various diseases. Neuropeptides, recognized as common biochemical mediators of communication between these systems, are receiving increasing attention because of their potential therapeutic applications in immune-related disorders. Additionally, many neuropeptides share significant similarities with antimicrobial peptides (AMPs), and evidence shows that these antimicrobial neuropeptides are directly involved in innate immunity. This review examines 10 antimicrobial neuropeptides, including pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), α-melanocyte stimulating hormone (α-MSH), ghrelin, adrenomedullin (AM), neuropeptide Y (NPY), urocortin II (UCN II), calcitonin gene-related peptide (CGRP), substance P (SP), and catestatin (CST). Their expression characteristics and the immunomodulatory mechanisms mediated by their specific receptors are summarized, along with potential drugs targeting these receptors. Future studies should focus on further investigating antimicrobial neuropeptides and advancing the development of related drugs in preclinical and/or clinical studies to improve the treatment of immune-related diseases.
Collapse
Affiliation(s)
- Kaiqi Chen
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| | - Xiaojun Wu
- College of Medical Engineering, Jining Medical University, Jining 272067, China; (X.W.); (R.L.)
| | - Xiaoke Li
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| | - Haoxuan Pan
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| | - Wenhui Zhang
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| | - Jinxi Shang
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| | - Yinuo Di
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| | - Ruonan Liu
- College of Medical Engineering, Jining Medical University, Jining 272067, China; (X.W.); (R.L.)
| | - Zhaodi Zheng
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| | - Xitan Hou
- College of Medical Imaging and Laboratory, Jining Medical University, Jining 272067, China; (K.C.); (X.L.); (H.P.); (W.Z.); (J.S.); (Y.D.)
| |
Collapse
|
49
|
Li Y, Liu Y, Gou M. Peptide with Dual Roles in Immune and Metabolic Regulation: Liver-Expressed Antimicrobial Peptide-2 (LEAP-2). Molecules 2025; 30:429. [PMID: 39860298 PMCID: PMC11767564 DOI: 10.3390/molecules30020429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP-2) was originally discovered as an antimicrobial peptide that plays a vital role in the host innate immune system of various vertebrates. Recent research discovered LEAP-2 as an endogenous antagonist and inverse agonist of the GHSR1a receptor. By acting as a competitive antagonist to ghrelin, LEAP-2 influences energy balance and metabolic processes via the ghrelin-GHSR1a signaling pathway. LEAP-2 alone or the LEAP-2/ghrelin molar ratio showed potential as therapeutic targets for obesity, diabetes, and metabolic disorders. This review explores the recent advances of LEAP-2 in immune modulation and energy regulation, highlighting its potential in treating the above diseases.
Collapse
Affiliation(s)
- Yitong Li
- College of Life Science, Liaoning Normal University, Dalian 116081, China;
- Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| | - Ying Liu
- Haixia Institute of Science and Technology, College of Horticulture, Fujian Agriculture and Forestry University, Fuzhou 350007, China;
| | - Meng Gou
- College of Life Science, Liaoning Normal University, Dalian 116081, China;
- Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| |
Collapse
|
50
|
Duan Z, Huang Y, Liu X, Tang H, Jiang G, Huang W, Du R, Zhou D, Yang H. A lower atherogenic index of plasma was associated with a higher incidence of sarcopenia. Sci Rep 2025; 15:2237. [PMID: 39825028 PMCID: PMC11742044 DOI: 10.1038/s41598-025-86398-w] [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: 10/28/2024] [Accepted: 01/10/2025] [Indexed: 01/20/2025] Open
Abstract
Sarcopenia is an age-related muscle senescence disease that leads to functional limitations, physical disability and premature death in older adults. Atherogenic index of plasma (AIP) is a novel indicator of atherosclerotic status based on triglycerides and high-density lipoprotein cholesterol. The aim of this study was to investigate the association between AIP and new-onset sarcopenia and its components among middle-aged and older adults in a Chinese community. This cohort study included 7,992 participants who were free of sarcopenia in 2011 in the China Health and Retirement Longitudinal Study and were followed up in 2013 and 2015. Sarcopenia was assessed using the recommendations of the Asian Working Group for Sarcopenia 2019. Longitudinal associations between AIP and sarcopenia and its components were assessed using Cox proportional risk regression modeling. The results showed that AIP was negatively associated with sarcopenia [HR and 95% CI: 0.73 (0.62-0.86)]; with muscle mass [β and 95%CI: 0.49 (0.4-0.57)], skeletal muscle mass index [β and 95%CI: 0.17 (0.15-0.2)], and grip strength [β and 95% CI: 0.17 (0.15-0.2)] being positively correlated. A lower AIP was associated with a lower muscle mass and handgrip strength and higher incidence of sarcopenia. Regular measurement of AIP in the middle-aged and older population in the community can help in the early diagnosis and intervention of sarcopenia.
Collapse
Affiliation(s)
- Zhiping Duan
- Department of Geriatrics, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, 292 Beijing Road, Kunming, 650011, Yunnan Province, China
| | - Yunda Huang
- Department of Geriatrics, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, 292 Beijing Road, Kunming, 650011, Yunnan Province, China
| | - Xiaoling Liu
- Radiotherapy Department, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Huiyu Tang
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Guihua Jiang
- Department of Geriatrics, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, 292 Beijing Road, Kunming, 650011, Yunnan Province, China
| | - Wei Huang
- Department of Geriatrics, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, 292 Beijing Road, Kunming, 650011, Yunnan Province, China
| | - Runfen Du
- Department of Geriatrics, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, 292 Beijing Road, Kunming, 650011, Yunnan Province, China
| | - Dan Zhou
- Mental Hospital of Yunnan Province, Mental Health Centre affiliated to Kunming Medical University, Mental Health Prevention and Control Centre of Yunnan Province, No. 733, Chuanjin Road, Kunming, 650224, Yunnan Province, China.
| | - Hong Yang
- Department of Geriatrics, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, 292 Beijing Road, Kunming, 650011, Yunnan Province, China.
| |
Collapse
|