1
|
Peteláková M, Neprašová B, Šmotková Z, Myšková A, Holá L, Petelák A, Áčová A, Cantel S, Fehrentz JA, Sýkora D, Kuneš J, Železná B, Maletínská L. Simultaneous treatment with palm-LEAP2(1-14) and feeding high-fat diet attenuates liver lipid metabolism but not obesity: Sign of selective resistance to palm-LEAP2(1-14). Mol Cell Endocrinol 2025; 597:112442. [PMID: 39689753 DOI: 10.1016/j.mce.2024.112442] [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: 08/18/2024] [Revised: 11/01/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Liver-enriched antimicrobial peptide 2 (LEAP2) is a natural antagonist/inverse agonist of ghrelin receptor GHSR. Its truncated palmitoylated analog palm-LEAP2(1-14) promised anti-obesity properties because it exhibited favourable stability and an acute anorexigenic effect in our previous studies. Here we demonstrate desirable palm-LEAP2(1-14) pharmacokinetics, with significant levels of the peptide persisting in mouse blood 3 h after its subcutaneous administration. Palm-LEAP2 (1-14) reduced ghrelin-induced c-Fos immunoreactivity in arcuate nucleus and area postrema, in line with previously described silencing of ghrelin orexigenic effect. In spite of this, anti-obesity effect was not reached by two-week palm-LEAP2(1-14) treatment in mice with diet-induced obesity. Similarly, palm-LEAP2(1-14) administered simultaneously with high-fat diet feeding for 8 weeks did not protect mice from development of obesity and related biochemical changes. However, palm-LEAP2(1-14) kept its ability to attenuate liver de novo lipogenesis, and prominently lowered liver gene expression of nuclear receptors PPARG, SREBF1 and PPARA, and also expression of lipogenic and lipolytic enzymes. In our recent study, we described a high-fat diet-induced ghrelin resistance, reversible by switch to standard diet, followed by resistance to the acute anorexigenic effects of palm-LEAP2(1-14). Here we conclude that this resistance to palm-LEAP2(1-14) in obesity is probably selective and does not concern its ability to inhibit liver lipid metabolism.
Collapse
Affiliation(s)
- Martina Peteláková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Barbora Neprašová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Šmotková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic; Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Aneta Myšková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic; University of Chemistry and Technology, Prague, Czech Republic
| | - Lucie Holá
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Petelák
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Áčová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Sonia Cantel
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - David Sýkora
- University of Chemistry and Technology, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic; Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
2
|
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
|
3
|
Perelló M. Critical Insights Into LEAP2 Biology and Physiological Functions: Potential Roles Beyond Ghrelin Antagonism. Endocrinology 2025; 166:bqaf011. [PMID: 39823403 DOI: 10.1210/endocr/bqaf011] [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: 10/01/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 01/19/2025]
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP2) has recently emerged as a novel hormone that reduces food intake and glycemia by acting through the growth hormone secretagogue receptor (GHSR), also known as the ghrelin receptor. This discovery has led to a fundamental reconceptualization of GHSR's functional dynamics, now understood to be under a dual and opposing regulation. LEAP2 exhibits several distinctive features. LEAP2 is released by hepatocytes and enterocytes, 2 cell types that lack classical regulatory secretory mechanisms and may respond differently to nutrient signals. LEAP2 is also found in higher concentrations in plasma than ghrelin, even under energy deficit conditions, and modulates GHSR by inhibiting both ghrelin-dependent and ghrelin-independent activities. Given these characteristics, LEAP2 appears to play a major role in regulating GHSR activity in vivo, extending beyond simple ghrelin antagonism and being crucial for the long-term regulation of energy balance. A deeper understanding of how LEAP2 functions may clarify the functional implications of GHSR in different physiological contexts and unlock new therapeutic strategies for treating obesity, diabetes, and other metabolic disorders.
Collapse
Affiliation(s)
- Mario Perelló
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE) (Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de La Plata, Comisión de Investigaciones Científicas de la Provincia de Buenos Aires), La Plata, Buenos Aires 1900, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, 751 24 Uppsala, Sweden
| |
Collapse
|
4
|
Sosinski O, Pruszynska-Oszmalek E, Leciejewska N, Sassek M, Kolodziejski PA. LEAP2 in Physiology-A Narrative Review. Int J Mol Sci 2025; 26:377. [PMID: 39796232 PMCID: PMC11722547 DOI: 10.3390/ijms26010377] [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: 10/23/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Liver Enriched Antimicrobial Peptide 2 (LEAP2) is a fascinating peptide that has gained significant attention since its discovery in 2003. Initially identified as an antimicrobial peptide, LEAP2 has more recently been found to play a key role in the regulation of energy metabolism. One of the most notable functions of LEAP2 is its interaction with the ghrelin hormone, which is known for stimulating hunger. LEAP2 acts as an inhibitor of ghrelin, thereby reducing food intake and influencing energy balance. The physiological roles of LEAP2 extend beyond appetite suppression. Studies have shown that LEAP2 has an impact on insulin secretion, suggesting its potential involvement in glucose metabolism and possibly insulin sensitivity, which is crucial in managing conditions like type 2 diabetes. Moreover, LEAP2 levels appear to fluctuate based on factors such as gender, developmental stage, and even interventions like bariatric surgery, which is known for its role in managing obesity and diabetes. Given these findings, LEAP2 shows potential as a therapeutic target, particularly for addressing obesity and metabolic diseases such as type 2 diabetes. Its ability to influence food intake and energy balance makes it a promising candidate for further research into therapies aimed at weight regulation and glycemic control. In the future, LEAP2 could become an important agent in the development of treatments aimed at curbing obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
| | - Ewa Pruszynska-Oszmalek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35 Street, 60-637 Poznan, Poland; (O.S.); (N.L.); (M.S.)
| | | | | | - Pawel Antoni Kolodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35 Street, 60-637 Poznan, Poland; (O.S.); (N.L.); (M.S.)
| |
Collapse
|
5
|
Emini M, Bhargava R, Aldhwayan M, Chhina N, Rodriguez Flores M, Aldubaikhi G, Al Lababidi M, Al-Najim W, Miras AD, Ruban A, Glaysher MA, Prechtl CG, Byrne JP, Teare JP, Goldstone AP. Satiety Hormone LEAP2 After Low-Calorie Diet With/Without Endobarrier Insertion in Obesity and Type 2 Diabetes Mellitus. J Endocr Soc 2024; 9:bvae214. [PMID: 39659543 PMCID: PMC11631353 DOI: 10.1210/jendso/bvae214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Indexed: 12/12/2024] Open
Abstract
Context The liver/foregut satiety hormone liver-expressed antimicrobial peptide 2 (LEAP2) is an inverse agonist at the acyl ghrelin receptor (GHSR), increasing after food intake and decreasing after bariatric surgery and short-term nonsurgical weight loss, but effects of long-term dietary weight loss are unknown. Objective The objective of this study was to examine and compare the effects of these interventions on fasting and postprandial plasma LEAP2 and investigate potential metabolic mediators of changes in plasma LEAP2. Methods Plasma LEAP2 was measured in a previously published 2-year trial comparing standard medical management (SMM) (including 600-kcal/day deficit) with duodenal-jejunal bypass liner (DJBL, Endobarrier) insertion (explanted after 1 year) in adults with obesity and inadequately controlled type 2 diabetes mellitus. Results In the SMM group (n = 25-37), weight decreased by 4.3%, 8.1%, 7.8%, and 6.4% at 2, 26, 50, and 104 weeks and fasting plasma LEAP2 decreased from baseline mean ± SD 15.3 ± 0.9 ng/mL by 1.7, 3.8, 2.1, and 2.0 ng/mL, respectively. Absolute/decreases in fasting plasma LEAP2 positively correlated with absolute/decreases in body mass index, glycated hemoglobin A1c, fasting plasma glucose, serum insulin, homeostatic model assessment for insulin resistance, and serum triglycerides. Despite greater weight loss in the DJBL group (n = 23-30) at 26 to 50 weeks (10.4%-11.4%), the decrease in fasting plasma LEAP2 was delayed and attenuated (vs SMM), which may contribute to greater weight loss by attenuating GHSR signaling. Plasma LEAP2 did not increase with weight regain from 50 to 104 weeks after DJBL explant, suggesting a new set point with weight loss maintenance. Increases in plasma LEAP2 after a 600-kcal meal (10.8%-16.1% at 1-2 hours) were unaffected by weight loss, improved glucose metabolism, or DJBL insertion (n = 9-25), suggesting liver rather than duodenum/jejunum may be the primary source of postprandial LEAP2 secretion. Conclusion These findings add to our understanding of the regulation and potential physiological role of plasma LEAP2.
Collapse
Affiliation(s)
- Mimoza Emini
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Raghav Bhargava
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Madhawi Aldhwayan
- College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Navpreet Chhina
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Marcela Rodriguez Flores
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Ghadah Aldubaikhi
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Moaz Al Lababidi
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Werd Al-Najim
- Department of Metabolism, Diabetes and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Alexander D Miras
- Department of Metabolism, Diabetes and Reproduction, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Aruchuna Ruban
- Department of Surgery and Cancer, Imperial College London, St. Mary‘s Hospital, London W2 1NY, UK
| | - Michael A Glaysher
- Division of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Christina G Prechtl
- Clinical Trials Unit, Department of Public Health, Imperial College London, London W12 7TA, UK
| | - James P Byrne
- Division of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Julian P Teare
- Department of Surgery and Cancer, Imperial College London, St. Mary‘s Hospital, London W2 1NY, UK
| | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
6
|
Freij K, Cleveland B, Biga P. Remodeling of the epigenetic landscape in rainbow trout, Oncorhynchus mykiss, offspring in response to maternal choline intake. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101348. [PMID: 39515277 DOI: 10.1016/j.cbd.2024.101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
This project focused on evaluating the effects of maternal dietary choline intake on global DNA methylation profiles and related transcriptional changes in rainbow trout offspring. Three experimental diets were formulated to test different levels of choline intake: (a) 2065 ppm choline (Low Choline, 0 % supplementation), (b) 5657 ppm choline (Medium Choline, 0.6 % supplementation), and (c) 9248 ppm choline (High Choline, 1.2 % choline supplementation). Six rainbow trout families were fed experimental diets beginning 18 months post-hatch until spawning; their offspring were fed a commercial diet. Reduced representation bisulfite sequencing (RRBS) was utilized to measure genome-wide methylation in offspring immediately after hatching. When comparing to the Medium Choline offspring, differential DNA methylation occurred more in the Low Choline offspring than High Choline, especially in genic features like promoters. The differentially methylated CpGs (q ≤ 0.01) were identified evenly between CpG islands and shores in the genome, mostly found in the introns of genes. Genes such as fabp2 and leap2B associated with protein binding, fatty acid binding, DNA binding, and response to bacteria were differentially methylated and detected as differentially regulated genes by previous RNA-seq analysis. Although these findings indicate that levels of dietary choline available in broodstock diets alter offspring DNA methylation;, most differentially expressed genes were not associated with differential DNA methylation, suggesting additional mechanisms playing a role in regulating gene expression in response to maternal choline intake.
Collapse
Affiliation(s)
- Khalid Freij
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA. https://twitter.com/@FreijKhalid
| | - Beth Cleveland
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service (ARS-USDA), Kearneysville, WV 25430, USA
| | - Peggy Biga
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
7
|
Islam MN, Nabekura H, Ueno H, Nishida T, Nanashima A, Sakoda H, Zhang W, Nakazato M. Liver-expressed antimicrobial peptide 2 is a hepatokine regulated by ghrelin, nutrients, and body weight. Sci Rep 2024; 14:24782. [PMID: 39433849 PMCID: PMC11494003 DOI: 10.1038/s41598-024-74048-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP2) is a peptide that counteracts the hunger hormone ghrelin-induced functions. Recently, we showed that vertical sleeve gastrectomy (VSG) did not alter the serum LEAP2 concentration in individuals with obesity. Here, we investigated the effects of VSG in both chow diet (CD)-fed and high-fat diet (HFD)-fed mice. In CD-fed mice, VSG increased plasma LEAP2 levels and hepatic Leap2 mRNA levels while decreasing body weight, blood glucose levels, and ghrelin levels. Intraperitoneal (ip) administration of ghrelin reversed these changes. These effects were found in both male and female mice. In contrast, VSG or weight loss in HFD-induced obese mice decreased LEAP2 levels. After fasting, the plasma LEAP2 concentration was in the following order: hepatic vein > abdominal aorta > portal vein. A high glucose concentration robustly increased the plasma LEAP2 concentration in the hepatic vein and abdominal aorta but not in the portal vein. In addition, corn oil or palmitate increased LEAP2 expression and secretion. The increase in LEAP2 levels after the meal tolerance test was delayed in the human subjects with diabetes. Our data suggest that various factors (metabolic, hormonal, and nutritional) regulate LEAP2, and the liver is the predominant site for the production and secretion of LEAP2. Furthermore, the interaction between ghrelin and LEAP2 is involved in the pathogenesis of obesity and diabetes.
Collapse
Affiliation(s)
- Md Nurul Islam
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hiroki Nabekura
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Division of Haematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hiroaki Ueno
- Division of Haematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takahiro Nishida
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Atsushi Nanashima
- Division of Hepato-Biliary-Pancreas Surgery, Department of Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hideyuki Sakoda
- Laboratory of Biomolecular Analysis, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Weidong Zhang
- Laboratory of Biomolecular Analysis, Institute for Protein Research, Osaka University, Osaka, Japan
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Masamitsu Nakazato
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyamacho, Toyonaka-shi, Osaka, Japan.
| |
Collapse
|
8
|
Tufvesson-Alm M, Aranäs C, Blid Sköldheden S, Vestlund J, Edvardsson CE, Jerlhag E. LEAP2, a ghrelin receptor inverse agonist, and its effect on alcohol-related responses in rodents. Transl Psychiatry 2024; 14:401. [PMID: 39358354 PMCID: PMC11446955 DOI: 10.1038/s41398-024-03136-y] [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: 06/11/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
The underlying neurobiology of alcohol use disorder (AUD) is complex and needs further unraveling, with one of the key mechanisms being the gut-brain peptide ghrelin and its receptor (GHSR). However, additional substrates of the ghrelin pathway, such as liver-expressed antimicrobial peptide 2 (LEAP2), an endogenous GHSR inverse agonist, may contribute to this neurobiological framework. While LEAP2 modulates feeding and reward through central mechanisms, its effects on alcohol responses are unknown. The aim of the present study was therefore to identify the impact of central LEAP2 on the ability of alcohol to activate the mesolimbic dopamine system and to define its ability to control alcohol intake. These experiments revealed that central LEAP2 (i.e. into the third ventricle) prevented the ability of alcohol to cause locomotor stimulation in male mice, suppressed the memory of alcohol reward and attenuated the dopamine release in the nucleus accumbens caused by alcohol. Moreover, central LEAP2 reduced alcohol consumption in both male and female rats exposed to alcohol for 6 weeks before treatment. However, the serum levels of LEAP2 were similar between high- and low- alcohol-consuming (male) rats. Furthermore, central LEAP2 lowered the food intake in the alcohol-consuming male rats and reduced the body weight in the females. Collectively, the present study revealed that central LEAP2 mitigates alcohol-related responses in rodents, contributing to our understanding of the ghrelin pathway's role in alcohol effects.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cajsa Aranäs
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Blid Sköldheden
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jesper Vestlund
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christian E Edvardsson
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
9
|
Guan K, Ye M, Guo A, Chen X, Shan Y, Li X. Deficiency of leap2 promotes somatic growth in zebrafish: Involvement of the growth hormone system. Heliyon 2024; 10:e36397. [PMID: 39347412 PMCID: PMC11437977 DOI: 10.1016/j.heliyon.2024.e36397] [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: 05/24/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 10/01/2024] Open
Abstract
Purpose Liver-expressed antimicrobial peptide-2 (LEAP2) is identified as an endogenous antagonist and inverse agonist of the growth hormone secretagogue receptor type 1a (GHSR1a), its effect on the GHSR1a is contrary to the role of GHRELIN. Growth hormone (GH) is a crucial hormone for early development. Previous studies report that LEAP2 dose-dependently attenuates ghrelin-induced GH secretion, and Leap2-knockout mice exhibit increased plasma GH levels after GHRELIN administration. Clinical data revealed a possible correlation between LEAP2 and height development. However, the role of LEAP2 in early development remains unclear. This study aimed to investigate the role of LEAP2 in early development using leap2 mutant zebrafish larvae as a model. Method We analyzed the conservation of LEAP2 peptide across multiple species and generated leap2 mutants in zebrafish by CRISPR-Cas9, dynamically observed and measured the growth and development of zebrafish larvae from fertilization to 5 day post fertilization (dpf). In situ hybridization, transcriptome sequencing, quantitative real-time PCR and Western blot were used to detect the expression levels of GH and its signaling in early stage of embryonic development. Result Our data demonstrate that zebrafish with a knockout of the leap2 gene display a significant increase in hatching rate, body length, and the distance between their eyes, all without visible developmental defects in the early stages of development. In addition, both RNA and protein analyses revealed a significant increase in GH expression in leap2 mutant. Conclusion In general, this study demonstrates that LEAP2 regulates the expression of GH during early development, particularly influencing body length.
Collapse
Affiliation(s)
- Kaiyu Guan
- Zhejiang Clinical Research Center for Mental Disorders, The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Minjie Ye
- Zhejiang Clinical Research Center for Mental Disorders, The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Anqi Guo
- Zhejiang Clinical Research Center for Mental Disorders, The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiaoyu Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yunfeng Shan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xi Li
- Zhejiang Clinical Research Center for Mental Disorders, The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
10
|
Varshney S, Shankar K, Kerr HL, Anderson LJ, Gupta D, Metzger NP, Singh O, Ogden SB, Paul S, Piñon F, Osborne-Lawrence S, Richard CP, Lawrence C, Mani BK, Garcia JM, Zigman JM. The LEAP2 Response to Cancer-Related Anorexia-Cachexia Syndrome in Male Mice and Patients. Endocrinology 2024; 165:bqae132. [PMID: 39331742 PMCID: PMC11481018 DOI: 10.1210/endocr/bqae132] [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: 06/06/2024] [Revised: 09/04/2024] [Accepted: 09/27/2024] [Indexed: 09/29/2024]
Abstract
The hormone ghrelin serves a protective role in cancer-related anorexia-cachexia syndrome (CACS)-a condition in which plasma levels of ghrelin rise, its administration lessens CACS severity, and experimentally reduced signaling by its receptor (GHSR) worsens fat loss and anorexia and accelerates death. Yet, actions for the related hormone liver-expressed antimicrobial peptide-2 (LEAP2), which is an endogenous GHSR antagonist, are unexplored in CACS. Here, we found that plasma LEAP2 and LEAP2/ghrelin ratio were lower in Lewis lung carcinoma (LLC) and RM-9 prostate cancer CACS mouse models. Ghrelin deletion exaggerated losses of tumor-free body weight and fat mass, reduced food intake, reduced soleus muscle weight, and/or lowered grip strength in LLC or RM-9 tumor-bearing mice. LEAP2 deletion lessened reductions in tumor-free body weight and fat mass and increased food intake in LLC or RM-9 tumor-bearing mice. In a 55-subject cohort of patients with CACS or weight-stable cancer, the plasma LEAP2/total ghrelin ratio was negatively correlated with 6-month weight change preceding blood collection. These data demonstrate that ghrelin deletion exacerbates CACS in the LLC and RM-9 tumor-bearing mouse models while contrastingly, LEAP2 deletion reduces measures of CACS in these tumor-bearing mouse models. Further, they suggest that lower plasma LEAP2/ghrelin ratio protects against worsened CACS.
Collapse
Affiliation(s)
- Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Haiming L Kerr
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Lindsey J Anderson
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Nathan P Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sean B Ogden
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Subhojit Paul
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Francisco Piñon
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Corine P Richard
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Connor Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Bharath K Mani
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jose M Garcia
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Division of Endocrinology & Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
11
|
Fei Y, Bao Z, Wang Q, Zhu Y, Lu J, Ouyang L, Hu Q, Zhou Y, Chen L. CRISPR/Cas9-induced LEAP2 and GHSR1a knockout mutant zebrafish displayed abnormal growth and impaired lipid metabolism. Gen Comp Endocrinol 2024; 355:114563. [PMID: 38830459 DOI: 10.1016/j.ygcen.2024.114563] [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: 01/04/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
Investigating the principles of fish fat deposition and conducting related research are current focal points in fish nutrition. This study explores the endocrine regulation of LEAP2 and GHSR1a in zebrafish by constructing mutantmodels andexamining the effects of the endocrine factors LEAP2 and its receptor GHSR1a on zebrafish growth, feeding, and liver fat deposition. Compared to the wild type (WT), the mutation of LEAP2 results in increased feeding and decreased swimming in zebrafish. The impact is more pronounced in adult female zebrafish, characterized by increased weight, length, width, and accumulation of lipid droplets in the liver.Incontrast, deficiency in GHSR1a significantly reduces the growth of male zebrafish and markedly decreases liver fat deposition.These research findings indicate the crucial roles of LEAP2 and GHSR1a in zebrafish feeding, growth, and intracellular fat metabolism. This study, for the first time, investigated the endocrine metabolic regulation functions of LEAP2 and GHSR1a in the model organism zebrafish, providing initial insights into their effects and potential mechanisms on zebrafish fat metabolism.
Collapse
Affiliation(s)
- Yueyue Fei
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Zhonggui Bao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Qin Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yihong Zhu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jigang Lu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Linyue Ouyang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Quiqin Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yan Zhou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Liangbiao Chen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
12
|
Jerlhag E. Ghrelin system and GLP-1 as potential treatment targets for alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:401-432. [PMID: 39523062 DOI: 10.1016/bs.irn.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Peptides of the gut-brain axis have gained recent attention as potential treatment targets for addiction. While the number of gut-brain peptides is vast, ghrelin and glucagon-like peptide-1 (GLP-1) have been suggested as important players. Ghrelin is traditionally considered an orexigenic peptide, but recent studies found that it increases alcohol intake in rodents and craving for alcohol in humans. Additionally, suppression of the ghrelin receptor attenuates alcohol-related responses in animal models reflecting alcohol use disorder (AUD). For instance, a lower alcohol intake, suppressed motivation to consume alcohol, and attenuated reward from alcohol is observed after ghrelin receptor antagonism treatment. On a similar note, a partial ghrelin receptor agonist prevents hangover symptoms in humans. When it comes to the anorexigenic peptide GLP-1, agonists of its receptor are approved to treat diabetes type 2 and obesity. Extensive preclinical studies have revealed that these GLP-1 receptor agonists reduce alcohol intake, suppress the motivation to consume alcohol, and prevent relapse drink, with effects tentatively associated with a reduced alcohol-induced reward. These preclinical findings have to some extent been varied in humans, as GLP-1 receptor agonists decrease alcohol intake in overweight patients with AUD. Furthermore, genetic variations in either the genes encoding for pre-pro-ghrelin, GHSR, GLP-1, or its receptor, are associated with AUD and heavy alcohol drinking. While central mechanisms appear to modulate the ability of either ghrelin or GLP-1 to regulate alcohol-related responses the exact mechanisms have not been defined. Taken together these preclinical and clinical data imply that gut-brain peptides participate in the addiction process and should be considered as potential targets for AUD treatment.
Collapse
Affiliation(s)
- Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
13
|
Englund A, Gilliam-Vigh H, Suppli MP, Gasbjerg LS, Vilsbøll T, Knop FK. Intestinal expression profiles and hepatic expression of LEAP2, ghrelin and their common receptor, GHSR, in humans. Peptides 2024; 177:171227. [PMID: 38657907 DOI: 10.1016/j.peptides.2024.171227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP2) and ghrelin have reciprocal effects on their common receptor, the growth hormone secretagogue receptor (GHSR). Ghrelin is considered a gastric hormone and LEAP2 a liver-derived hormone and both have been proposed to be involved in the pathophysiology of obesity and type 2 diabetes (T2D). We investigated the mRNA expression of LEAP2, ghrelin and GHSR along the intestinal tract of individuals with and without TD2, and in the liver of men with and without obesity. Mucosal biopsies retrieved with 30-cm intervals throughout the small intestine and from 7 well-defined locations along the large intestine from 12 individuals with T2D and 12 healthy controls together with liver biopsies from 15 men with obesity and 15 lean men were subjected to bulk transcriptomics analysis. Both in individuals with and without T2D, mRNA expression of LEAP2 increased through the small intestine until dropping at the ileocecal valve, with little LEAP2 mRNA expression in the large intestine. Pronounced LEAP2 expression was observed in the liver of men with and without obesity. Robust ghrelin mRNA expression was observed in the duodenum of individuals with and without T2D, gradually decreasing along the small intestine with little expression in the large intestine. Ghrelin mRNA expression was not detected in the liver biopsies, and GHSR mRNA expression was not. In conclusion, we provide unique mRNA expression profiles of LEAP2, ghrelin and GHSR along the human intestinal tract showing no T2D-associated changes, and in the liver showing no differences between men with and without obesity.
Collapse
Affiliation(s)
- Anders Englund
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
| | - Hannah Gilliam-Vigh
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
| | - Malte P Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
| | - Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark.
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark; Novo Nordisk A/S, Novo Allé, Bagsværd, Denmark.
| |
Collapse
|
14
|
Lin CE, Chen CY. Impacts of Central Administration of the Novel Peptide, LEAP-2, in Different Food Intake Models in Conscious Rats. Nutrients 2024; 16:1946. [PMID: 38931301 PMCID: PMC11206331 DOI: 10.3390/nu16121946] [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] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Liver-expressed antimicrobial peptide-2 (LEAP-2) has mutual antagonism with ghrelin, which evokes food intake under a freely fed state. Nevertheless, the impact of LEAP-2 on ghrelin under time-restricted feeding (TRF), which has benefits in the context of metabolic disease, is still unknown. This study aims to explore the impact of central administration of LEAP-2 on the ingestion behavior of rats, which was evaluated using their cumulative food intake in the TRF state. Before intracerebroventricular (ICV) administration of O-n-octanoylated ghrelin (0.1 nmol/rat), as a food-stimulatory model, the rats received various doses of LEAP-2 (0.3, 1, 3 nmol/rat, ICV). Cumulative food intake was recorded at 1, 2, 4, 8, 12, and 24 h after ICV injection under 12 h freely fed and TRF states in a light phase. In 12 h freely fed and TRF states, central administration of ghrelin alone induced feeding behavior. Pre-treatment with LEAP-2 (1 and 3 nmol/rat, ICV) suppressed ghrelin-induced food intake in a dose-dependent manner in a 12 h freely fed state instead of a TRF state, which may have disturbed the balance of ghrelin and LEAP-2. This study provides neuroendocrine-based evidence that may explain why TRF sometimes fails in fighting obesity/metabolic dysfunction-associated steatotic liver disease in clinics.
Collapse
Affiliation(s)
- Chia-En Lin
- Department of Pharmacy, Tajen University, No. 20, Weixin Rd., Yanpu Township, Pingtung County 907101, Taiwan;
| | - Chih-Yen Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Emergency and Critical Medicine, and School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Chinese Taipei Society for the Study of Obesity, Taipei 110301, Taiwan
| |
Collapse
|
15
|
Shankar K, Metzger NP, Lawrence C, Gupta D, Osborne-Lawrence S, Varshney S, Singh O, Richard CP, Zaykov AN, Rolfts R, DuBois BN, Perez-Tilve D, Mani BK, Hammer STG, Zigman JM. A long-acting LEAP2 analog reduces hepatic steatosis and inflammation and causes marked weight loss in mice. Mol Metab 2024; 84:101950. [PMID: 38697291 PMCID: PMC11103953 DOI: 10.1016/j.molmet.2024.101950] [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: 01/18/2024] [Revised: 04/01/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024] Open
Abstract
OBJECTIVE The number of individuals affected by metabolic dysfunction associated fatty liver disease [1] is on the rise, yet hormonal contributors to the condition remain incompletely described and only a single FDA-approved treatment is available. Some studies suggest that the hormones ghrelin and LEAP2, which act as agonist and antagonist/inverse agonist, respectively, for the G protein coupled receptor GHSR, may influence the development of MAFLD. For instance, ghrelin increases hepatic fat whereas synthetic GHSR antagonists do the opposite. Also, hepatic steatosis is less prominent in standard chow-fed ghrelin-KO mice but more prominent in 42% high-fat diet-fed female LEAP2-KO mice. METHODS Here, we sought to determine the therapeutic potential of a long-acting LEAP2 analog (LA-LEAP2) to treat MAFLD in mice. LEAP2-KO and wild-type littermate mice were fed a Gubra-Amylin-NASH (GAN) diet for 10 or 40 wks, with some randomized to an additional 28 or 10 days of GAN diet, respectively, while treated with LA-LEAP2 vs Vehicle. Various metabolic parameters were followed and biochemical and histological assessments of MAFLD were made. RESULTS Among the most notable metabolic effects, daily LA-LEAP2 administration to both LEAP2-KO and wild-type littermates during the final 4 wks of a 14 wk-long GAN diet challenge markedly reduced liver weight, hepatic triglycerides, plasma ALT, hepatic microvesicular steatosis, hepatic lobular inflammation, NASH activity scores, and prevalence of higher-grade fibrosis. These changes were accompanied by prominent reductions in body weight, without effects on food intake, and reduced plasma total cholesterol. Daily LA-LEAP2 administration during the final 10 d of a 41.5 wk-long GAN diet challenge also reduced body weight, plasma ALT, and plasma total cholesterol in LEAP2-KO and wild-type littermates and prevalence of higher grade fibrosis in LEAP2-KO mice. CONCLUSIONS Administration of LA-LEAP2 to mice fed a MAFLD-prone diet markedly improves several facets of MAFLD, including hepatic steatosis, hepatic lobular inflammation, higher-grade hepatic fibrosis, and transaminitis. These changes are accompanied by prominent reductions in body weight and lowered plasma total cholesterol. Taken together, these data suggest that LEAP2 analogs such as LA-LEAP2 hold promise for the treatment of MAFLD and obesity.
Collapse
Affiliation(s)
- Kripa Shankar
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Nathan P Metzger
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Connor Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Deepali Gupta
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Sherri Osborne-Lawrence
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Salil Varshney
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | - Corine P Richard
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA
| | | | - Rebecca Rolfts
- Novo Nordisk Lexington, 33 Hayden Ave, Lexington, MA 02421, USA
| | - Barent N DuBois
- Novo Nordisk Lexington, 33 Hayden Ave, Lexington, MA 02421, USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bharath K Mani
- Novo Nordisk Lexington, 33 Hayden Ave, Lexington, MA 02421, USA
| | - Suntrea T G Hammer
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9077, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, USA; Department of Psychiatry, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
16
|
Berthoud HR, Münzberg H, Morrison CD, Neuhuber WL. Hepatic interoception in health and disease. Auton Neurosci 2024; 253:103174. [PMID: 38579493 PMCID: PMC11129274 DOI: 10.1016/j.autneu.2024.103174] [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: 12/19/2023] [Revised: 03/14/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
The liver is a large organ with crucial functions in metabolism and immune defense, as well as blood homeostasis and detoxification, and it is clearly in bidirectional communication with the brain and rest of the body via both neural and humoral pathways. A host of neural sensory mechanisms have been proposed, but in contrast to the gut-brain axis, details for both the exact site and molecular signaling steps of their peripheral transduction mechanisms are generally lacking. Similarly, knowledge about function-specific sensory and motor components of both vagal and spinal access pathways to the hepatic parenchyma is missing. Lack of progress largely owes to controversies regarding selectivity of vagal access pathways and extent of hepatocyte innervation. In contrast, there is considerable evidence for glucose sensors in the wall of the hepatic portal vein and their importance for glucose handling by the liver and the brain and the systemic response to hypoglycemia. As liver diseases are on the rise globally, and there are intriguing associations between liver diseases and mental illnesses, it will be important to further dissect and identify both neural and humoral pathways that mediate hepatocyte-specific signals to relevant brain areas. The question of whether and how sensations from the liver contribute to interoceptive self-awareness has not yet been explored.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Christopher D Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Winfried L Neuhuber
- Institute for Anatomy and Cell Biology, Friedrich-Alexander University, Erlangen, Germany.
| |
Collapse
|
17
|
Mahalingam S, Bellamkonda R, Kharbanda KK, Arumugam MK, Kumar V, Casey CA, Leggio L, Rasineni K. Role of ghrelin hormone in the development of alcohol-associated liver disease. Biomed Pharmacother 2024; 174:116595. [PMID: 38640709 PMCID: PMC11161137 DOI: 10.1016/j.biopha.2024.116595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
Fatty liver is the earliest response of the liver to excessive alcohol consumption. Previously we identified that chronic alcohol administration increases levels of stomach-derived hormone, ghrelin, which by reducing circulating insulin levels, ultimately contributes to the development of alcohol-associated liver disease (ALD). In addition, ghrelin directly promotes fat accumulation in hepatocytes by enhancing de novo lipogenesis. Other than promoting ALD, ghrelin is known to increase alcohol craving and intake. In this study, we used a ghrelin receptor (GHSR) knockout (KO) rat model to characterize the specific contribution of ghrelin in the development of ALD with emphasis on energy homeostasis. Male Wistar wild type (WT) and GHSR-KO rats were pair-fed the Lieber-DeCarli control or ethanol diet for 6 weeks. At the end of the feeding period, glucose tolerance test was conducted, and tissue samples were collected. We observed reduced alcohol intake by GHSR-KOs compared to a previous study where WT rats were fed ethanol diet ad libitum. Further, when the WTs were pair-fed to GHSR-KOs, the KO rats exhibited resistance to develop ALD through improving insulin secretion/sensitivity to reduce adipose lipolysis and hepatic fatty acid uptake/synthesis and increase fatty acid oxidation. Furthermore, proteomic data revealed that ethanol-fed KO exhibit less alcohol-induced mitochondrial dysfunction and oxidative stress than WT rats. Proteomic data also confirmed that the ethanol-fed KOs are insulin sensitive and are resistant to hepatic steatosis development compared to WT rats. Together, these data confirm that inhibiting ghrelin action prevent alcohol-induced liver and adipose dysfunction independent of reducing alcohol intake.
Collapse
Affiliation(s)
- Sundararajan Mahalingam
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ramesh Bellamkonda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Madan Kumar Arumugam
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vikas Kumar
- Mass Spectrometry and Proteomic Core Facility, University of Nebraska Medical Center, Omaha, NE, USA; Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Carol A Casey
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Bethesda, Baltimore, MD, USA; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA; Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
18
|
Tufvesson-Alm M, Zhang Q, Aranäs C, Blid Sköldheden S, Edvardsson CE, Jerlhag E. Decoding the influence of central LEAP2 on food intake and its effect on accumbal dopamine release. Prog Neurobiol 2024; 236:102615. [PMID: 38641041 DOI: 10.1016/j.pneurobio.2024.102615] [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: 11/09/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
The gut-brain peptide ghrelin and its receptor are established as a regulator of hunger and reward-processing. However, the recently recognized ghrelin receptor inverse agonist, liver-expressed antimicrobial peptide 2 (LEAP2), is less characterized. The present study aimed to elucidate LEAP2s central effect on reward-related behaviors through feeding and its mechanism. LEAP2 was administrated centrally in mice and effectively reduced feeding and intake of palatable foods. Strikingly, LEAP2s effect on feeding was correlated to the preference of the palatable food. Further, LEAP2 reduced the rewarding memory of high preference foods, and attenuated the accumbal dopamine release associated with palatable food exposure and eating. Interestingly, LEAP2 was widely expressed in the brain, and particularly in reward-related brain areas such as the laterodorsal tegmental area (LDTg). This expression was markedly altered when allowed free access to palatable foods. Accordingly, infusion of LEAP2 into LDTg was sufficient to transiently reduce acute palatable food intake. Taken together, the present results show that central LEAP2 has a profound effect on dopaminergic reward signaling associated with food and affects several aspects of feeding. The present study highlights LEAP2s effect on reward, which may have applications for obesity and other reward-related psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Qian Zhang
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Cajsa Aranäs
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Sebastian Blid Sköldheden
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Christian E Edvardsson
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 13A, Gothenburg SE-405 30, Sweden.
| |
Collapse
|
19
|
Andreoli MF, Fittipaldi AS, Castrogiovanni D, De Francesco PN, Valdivia S, Heredia F, Ribet-Travers C, Mendez I, Fasano MV, Schioth HB, Doi SA, Habib AM, Perello M. Pre-prandial plasma liver-expressed antimicrobial peptide 2 (LEAP2) concentration in humans is inversely associated with hunger sensation in a ghrelin independent manner. Eur J Nutr 2024; 63:751-762. [PMID: 38157050 DOI: 10.1007/s00394-023-03304-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE The liver-expressed antimicrobial peptide 2 (LEAP2) is a newly recognized peptide hormone that acts via the growth hormone secretagogue receptor (GHSR) blunting the effects of ghrelin and displaying ghrelin-independent actions. Since the implications of LEAP2 are beginning to be elucidated, we investigated if plasma LEAP2 concentration varies with feeding status or sex and whether it is associated with glucose metabolism and appetite sensations. METHODS We performed a single test meal study, in which plasma concentrations of LEAP2, ghrelin, insulin and glucose as well as visual analogue scales for hunger, desire to eat, prospective food consumption, fullness were assessed before and 60 min after breakfast in 44 participants (n = 21 females) with normal weight (NW) or overweight/obesity (OW/OB). RESULTS Pre-prandial plasma LEAP2 concentration was ~ 1.6-fold higher whereas ghrelin was ~ 2.0-fold lower in individuals with OW/OB (p < 0.001) independently of sex. After adjusting for body mass index (BMI) and sex, pre-prandial plasma LEAP2 concentration displayed a direct relationship with BMI (β: 0.09; 95%CI: 0.05, 0.13; p < 0.001), fat mass (β: 0.05; 95%CI: 0.01, 0.09; p = 0.010) and glycemia (β: 0.24; 95%CI: 0.05, 0.43; p = 0.021), whereas plasma ghrelin concentration displayed an inverse relationship with BMI and fat mass but not with glycemia. Postprandial plasma LEAP2 concentration increased ~ 58% in females with OW/OB (p = 0.045) but not in females with NW or in males. Pre-prandial plasma LEAP2 concentration displayed an inverse relationship with hunger score (β: - 11.16; 95% CI: - 18.52, - 3.79; p = 0.004), in a BMI-, sex- and ghrelin-independent manner. CONCLUSIONS LEAP2 emerges as a key hormone implicated in the regulation of metabolism and appetite in humans. TRIAL REGISTRATION The study was retrospectively registered in clinicaltrials.gov (April 2023). CLINICALTRIALS gov Identifier: NCT05815641.
Collapse
Affiliation(s)
- María F Andreoli
- Instituto de Desarrollo e Investigaciones Pediátricas (IDIP), HIAEP Sor María Ludovica de la Plata, Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), Calle 63 # 1069, La Plata, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| | - Antonela S Fittipaldi
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE). Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y CIC-PBA, Calle 526 S/N Entre 10 y 11, La Plata, Buenos Aires, Argentina
| | - Daniel Castrogiovanni
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE). Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y CIC-PBA, Calle 526 S/N Entre 10 y 11, La Plata, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE). Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y CIC-PBA, Calle 526 S/N Entre 10 y 11, La Plata, Buenos Aires, Argentina
| | - Spring Valdivia
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE). Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y CIC-PBA, Calle 526 S/N Entre 10 y 11, La Plata, Buenos Aires, Argentina
| | - Florencia Heredia
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE). Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y CIC-PBA, Calle 526 S/N Entre 10 y 11, La Plata, Buenos Aires, Argentina
| | | | - Ignacio Mendez
- Instituto de Desarrollo e Investigaciones Pediátricas (IDIP), HIAEP Sor María Ludovica de la Plata, Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), Calle 63 # 1069, La Plata, Buenos Aires, Argentina
| | - María V Fasano
- Instituto de Desarrollo e Investigaciones Pediátricas (IDIP), HIAEP Sor María Ludovica de la Plata, Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), Calle 63 # 1069, La Plata, Buenos Aires, Argentina
- Centro de Matemática la Plata, Facultad de Ciencias Exactas, UNLP/CIC-PBA, La Plata, Argentina
| | - Helgi B Schioth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Suhail A Doi
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mario Perello
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE). Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y CIC-PBA, Calle 526 S/N Entre 10 y 11, La Plata, Buenos Aires, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
20
|
Kulkarni SS, Singh O, Zigman JM. The intersection between ghrelin, metabolism and circadian rhythms. Nat Rev Endocrinol 2024; 20:228-238. [PMID: 38123819 PMCID: PMC11760189 DOI: 10.1038/s41574-023-00927-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Despite the growing popular interest in sleep and diet, many gaps exist in our scientific understanding of the interaction between circadian rhythms and metabolism. In this Review, we explore a promising, bidirectional role for ghrelin in mediating this interaction. Ghrelin both influences and is influenced by central and peripheral circadian systems. Specifically, we focus on how ghrelin impacts outputs of circadian rhythm, including neuronal activity, circulating growth hormone levels, locomotor activity and eating behaviour. We also consider the effects of circadian rhythms on ghrelin expression and the consequences of disrupted circadian patterns, such as shift work and jet lag, on ghrelin secretion. Our Review is aimed at both the casual reader interested in gaining more insight into the scientific context surrounding the trending topics of sleep and metabolism, as well as experienced scientists in the fields of ghrelin and circadian biology seeking inspiration and a comprehensive overview of how these fields are related.
Collapse
Affiliation(s)
- Soumya S Kulkarni
- Medical Scientist Training Program, UT Southwestern Medical Center, Dallas, TX, USA
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
21
|
Wang S, Wang D, Bai Y, Zheng G, Han Y, Wang L, Hu J, Zhu H, Bai Y. Expression of Toll-like receptors and host defence peptides in the cecum of chicken challenged with Eimeria tenella. Parasite Immunol 2024; 46:e13022. [PMID: 38384176 DOI: 10.1111/pim.13022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 02/23/2024]
Abstract
Chicken coccidiosis, caused by Eimeria protozoa, affects poultry farming. Toll-like receptors (TLRs) and host defence peptides (HDPs) help host innate immune responses to eliminate invading pathogens, but their roles in Eimeria tenella infection remain poorly understood. Herein, 14-day-old chickens were treated orally with 50,000 E. tenella oocysts and the cecum was dissected at different timepoints. mRNA expression of 10 chicken TLRs (chTLRs) and five HDPs was measured by quantitative real-time PCR. chTLR7 and chTLR15 were upregulated significantly at 3 h post-infection while other chTLRs were downregulated (p < .05). chTLR1a, chTLR1b, chTLR2b and chTLR4 peaked at 36 h post-infection, chTLR3, chTLR5 and chTLR15 peaked at 72 h post-infection and chTLR21 expression was highest among chTLRs, peaking at 48 h post-infection (p < 0.05). For HDPs, cathelicidin (CATH) 1 to 3 and B1 peaked at 48 h post-infection, liver-expressed antimicrobial peptide 2 peaked at 96 h post-infection, and CATH 2 expression was highest among HDPs. CATH2 and CATH3 were markedly upregulated at 3 h post-infection (p < .05). The results provide insight into innate immune molecules during E. tenella infection in chicken, and indicate that innate immune responses may mediate resistance to chicken coccidiosis.
Collapse
Affiliation(s)
- Song Wang
- Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Danni Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, China
| | - Yilin Bai
- School of Agricultural Science, Zhengzhou University, Zhengzhou, China
| | - Guijie Zheng
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, China
| | - Yanhui Han
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, China
| | - Huili Zhu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, China
| | - Yueyu Bai
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, China
| |
Collapse
|
22
|
Howe SL, Holdom CJ, McCombe PA, Henderson RD, Zigman JM, Ngo ST, Steyn FJ. Associations of postprandial ghrelin, liver-expressed antimicrobial peptide 2 and leptin levels with body composition, disease progression and survival in patients with amyotrophic lateral sclerosis. Eur J Neurol 2024; 31:e16052. [PMID: 37658515 PMCID: PMC10840749 DOI: 10.1111/ene.16052] [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: 06/03/2023] [Revised: 08/06/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND AND PURPOSE Loss of appetite contributes to weight loss and faster disease progression in amyotrophic lateral sclerosis (ALS). Impairment of appetite control in ALS may include altered production or action of orexigenic (i.e., ghrelin) and anorexigenic (i.e., liver-expressed antimicrobial peptide 2 [LEAP2] and leptin) hormones. We aimed to determine if postprandial circulating ghrelin levels, LEAP2 levels, LEAP2:ghrelin molar ratio and leptin levels differ in ALS patients compared to non-neurodegenerative disease controls, and whether they are associated with disease progression and body composition. METHODS In this prospective natural history study, we assessed postprandial plasma levels of ghrelin, LEAP2 and leptin in patients with ALS (cases; n = 46) and controls (controls; n = 43). For cases, measures were compared to changes in body weight, body composition and clinical outcomes. RESULTS Postprandial ghrelin level was decreased by 52% in cases compared to controls (p = 0.013). LEAP2:ghrelin molar ratio was increased by 249% (p = 0.009), suggesting greater ghrelin resistance. Patients with lower LEAP2:ghrelin tended to have better functional capacity at assessment, as inferred by the ALS Functional Rating Scale-Revised (τ = -0.179, p = 0.086). Furthermore, ghrelin and LEAP2:ghrelin molar ratio correlated with diagnostic delay (ghrelin, τ = 0.223, p = 0.029; LEAP2:ghrelin, τ = -0.213, p = 0.037). Baseline ghrelin level, LEAP2 level, LEAP2:ghrelin ratio and leptin level were, however, not predictive of change in functional capacity during follow-up. Also, patients with higher postprandial ghrelin levels (hazard ratio [HR] 1.375, p = 0.048), and lower LEAP2:ghelin ratios (HR 0.828, p = 0.051) had an increased risk of earlier death. CONCLUSIONS Reduced postprandial ghrelin levels, coupled with increased LEAP2:ghrelin molar ratios, suggests a loss of ghrelin action in patients with ALS. Given ghrelin's actions on appetite, metabolism and neuroprotection, reduced ghrelin and greater ghrelin resistance could contribute to impaired capacity to tolerate the physiological impact of disease. Comprehensive studies are needed to explain how ghrelin and LEAP2 contribute to body weight regulation and disease progression in ALS.
Collapse
Affiliation(s)
- Stephanie L. Howe
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
| | - Cory J. Holdom
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
| | - Pamela A. McCombe
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Robert D. Henderson
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Shyuan T. Ngo
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Frederik J. Steyn
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
- School of Biomedical SciencesThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
23
|
Pedersen MGB, Lauritzen ES, Svart MV, Støy J, Søndergaard E, Thomsen HH, Kampmann U, Bjerre M, Jessen N, Møller N, Rittig N. Nutrient sensing: LEAP2 concentration in response to fasting, glucose, lactate, and β-hydroxybutyrate in healthy young males. Am J Clin Nutr 2023; 118:1091-1098. [PMID: 37844838 DOI: 10.1016/j.ajcnut.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND The appetite-suppressing potential of liver-expressed antimicrobial peptide 2 (LEAP2), and its antagonistic effects on the hunger-inducing hormone ghrelin have attracted scientific interest. It is unclear how LEAP2 is influenced by fasting and how it responds to specific nutrients. OBJECTIVES The purpose of this investigation was to assess whether LEAP2 concentration 1) decreases after fasting, 2) increases postprandially, and 3) is regulated by nutrient sensing in the splanchnic bed. METHODS Plasma LEAP2 concentration was measured in blood samples from 5 clinical cross-over trials, following 1) 36 h of fasting (n = 8), 2) 10 h of fasting (n = 37, baseline data pooled from 4 of the clinical trials), 3) Oral and intravenous glucose administration (n = 11), 4) Oral and intravenous Na-lactate administration (n = 10), and 5) Oral and intravenous Na-β-hydroxybutyrate (BHB) administration (n = 8). All 5 trials included healthy males. RESULTS Compared with a 10-h fasting period, the median LEAP2 concentration was 38% lower following 36 h of fasting (P < 0.001). Oral administration of glucose elevated, whereas intravenous glucose administration lowered LEAP2 concentration (intervention x time, P = 0.001), resulting in a mean difference of 9 ng/mL (95% confidence interval [CI]: 1, 17) after 120 min. Oral lactate increased, and intravenous lactate decreased LEAP2 (intervention x time, P = 0.007), with a mean difference between interventions of 10 ng/mL (95% CI: 6, 15) after 120 min. In contrast, oral and intravenous administration of BHB reduced the LEAP2 concentration (main effect of time, P < 0.001). CONCLUSIONS Our investigations show that LEAP2 concentration was lower after a 36-h fast than an overnight fast and that oral delivery of glucose and lactate elevated LEAP2 concentration compared with intravenous administration, whereas LEAP2 concentrations decreased with both oral and intravenous BHB. This indicates that the LEAP2 concentration is sensitive to intestinal exposure to specific substrates, highlighting the need for future studies exploring the relationship between nutrients and LEAP2. This trial was registered at clinicaltrials.gov as NCT01840098, NCT03204877, NCT04299815, NCT03935841, and NCT01705782.
Collapse
Affiliation(s)
- Mette Glavind Bülow Pedersen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark; Medical/Steno Aarhus Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark.
| | | | - Mads Vandsted Svart
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark; Medical/Steno Aarhus Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Julie Støy
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark
| | - Esben Søndergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus C, Denmark
| | - Henrik Holm Thomsen
- Department of Internal Medicine, Viborg Regional Hospital, Viborg, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus C, Denmark
| | - Ulla Kampmann
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus C, Denmark
| | - Mette Bjerre
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark; Medical/Steno Aarhus Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark
| | - Niels Møller
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark; Medical/Steno Aarhus Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Nikolaj Rittig
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark; Medical/Steno Aarhus Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| |
Collapse
|
24
|
Holá L, Tureckiuová T, Kuneš J, Železná B, Maletínská L. High-Fat Diet Induces Resistance to Ghrelin and LEAP2 Peptide Analogs in Mice. Physiol Res 2023; 72:607-619. [PMID: 38015760 PMCID: PMC10751049 DOI: 10.33549/physiolres.935189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/01/2023] [Indexed: 01/05/2024] Open
Abstract
Recent data suggest that the orexigenic peptide ghrelin and liver-expressed antimicrobial peptide 2 (LEAP2) have opposing effects on food intake regulation. Although circulating ghrelin is decreased in obesity, peripheral ghrelin administration does not induce food intake in obese mice. Limited information is available on ghrelin resistance in relation to LEAP2. In this study, the interplay between ghrelin and LEAP2 in obesity induced by a high-fat (HF) diet in mice was studied. First, the progression of obesity and intolerance to glucose together with plasma levels of active and total ghrelin, leptin, as well as liver LEAP2 mRNA expression at different time points of HF diet feeding was examined. In addition, the impact of switch from a HF diet to a standard diet on plasma ghrelin and LEAP2 production was studied. Second, sensitivity to the stable ghrelin analogue [Dpr3]Ghrelin or our novel LEAP2 analogue palm-LEAP2(1-14) during the progression of HF diet-induced obesity and after the switch for standard diet was investigated. Food intake was monitored after acute subcutaneous administration. HF diet feeding decreased both active and total plasma ghrelin and increased liver LEAP2 mRNA expression along with intolerance to glucose and the switch to a standard diet normalized liver LEAP2 mRNA expression and plasma level of active ghrelin, but not of total ghrelin. Additionally, our study demonstrates that a HF diet causes resistance to [Dpr3]Ghrelin, reversible by switch to St diet, followed by resistance to palm-LEAP2(1-14). Further studies are needed to determine the long-term effects of LEAP2 analogues on obesity-related ghrelin resistance.
Collapse
Affiliation(s)
- L Holá
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Praha 6, Czech Republic.
| | | | | | | | | |
Collapse
|
25
|
Maric I, López-Ferreras L, Bhat Y, Asker M, Börchers S, Bellfy L, Byun S, Kwapis JL, Skibicka KP. From the stomach to locus coeruleus: new neural substrate for ghrelin's effects on ingestive, motivated and anxiety-like behaviors. Front Pharmacol 2023; 14:1286805. [PMID: 38026980 PMCID: PMC10679437 DOI: 10.3389/fphar.2023.1286805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Ghrelin, a stomach-derived orexigenic hormone, has a well-established role in energy homeostasis, food reward, and emotionality. Noradrenergic neurons of the locus coeruleus (LC) are known to play an important role in arousal, emotion, cognition, but recently have also been implicated in control of feeding behavior. Ghrelin receptors (the growth hormone secretagogue receptor, GHSR) may be found in the LC, but the behavioral effects of ghrelin signaling in this area are still unexplored. Here, we first determined whether GHSR are present in the rat LC, and demonstrate that GHSR are expressed on noradrenergic neurons in both sexes. We next investigated whether ghrelin controls ingestive and motivated behaviors as well as anxiety-like behavior by acting in the LC. To pursue this idea, we examined the effects of LC GHSR stimulation and blockade on food intake, operant responding for a palatable food reward and, anxiety-like behavior in the open field (OF) and acoustic startle response (ASR) tests in male and female rats. Our results demonstrate that intra-LC ghrelin administration increases chow intake and motivated behavior for sucrose in both sexes. Additionally, females, but not males, exhibited a potent anxiolytic response in the ASR. In order to determine whether activation of GHSR in the LC was necessary for feeding and anxiety behavior control, we utilized liver-expressed antimicrobial peptide 2 (LEAP2), a newly identified endogenous GHSR antagonist. LEAP2 delivered specifically into the LC was sufficient to reduce fasting-induced chow hyperphagia in both sexes, but food reward only in females. Moreover, blockade of GHSR in the LC increased anxiety-like behavior measured in the ASR test in both sexes. Taken together, these results indicate that ghrelin acts in the LC to alter ingestive, motivated and anxiety-like behaviors, with a degree of sex divergence.
Collapse
Affiliation(s)
- Ivana Maric
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Lorena López-Ferreras
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yashaswini Bhat
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Mohammed Asker
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Stina Börchers
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Lauren Bellfy
- Department of Biology, Pennsylvania State University, State College, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, PA, United States
| | - Suyeun Byun
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
| | - Janine L. Kwapis
- Department of Biology, Pennsylvania State University, State College, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, PA, United States
| | - Karolina P. Skibicka
- Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, State College, PA, United States
| |
Collapse
|
26
|
Beheshti S, Ershadi S, Zamani F, Azimzadeh M, Wesal MW. Differential impact of a ghrelin receptor antagonist or inverse agonist in the electrical kindling model of epilepsy. Epilepsy Res 2023; 197:107234. [PMID: 37793283 DOI: 10.1016/j.eplepsyres.2023.107234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/06/2023]
Abstract
Ghrelin is a peptide, which has been shown to affect seizures. However, there is not a consensus about its real impact on the control of seizure severity. We assessed the influence of intra-amygdala injections of a ghrelin receptor (GHSR) antagonist, as well as a GHSR inverse agonist on the electrical kindling-induced seizures. Two unipolar electrodes and a tripolar electrode twisted with a guide cannula were implanted in the skull surface or the basolateral amygdala of adult male rats, respectively. A rapid electrical kindling protocol was applied for kindling epileptogenesis. The stimulations were applied until rats showed three consecutive stage five seizures. Each rat was considered as its control. D-Lys-3-GHRP-6 (1, 12.5, and 25 μg/rat) or [D-Arg, D-phe, D-Trp, heu] substance P (D-SP) (50, 500 and 5000 ng/rat) as the GHSR antagonist or inverse agonist were injected into the basolateral amygdala. Seizure parameters including after-discharge duration (ADD), stage five duration (S5D), and seizure stage (SS) were documented thirty minutes following administration of the drugs or saline. Antagonism of the GHSR in the amygdala, significantly increased seizure induction in the kindled rats, in a dose-dependent manner, and induced spontaneous seizures leading to status epilepticus. Conversely, D-SP had a dose-dependent anticonvulsant activity, indicated by decreased ADD and S5D. The results show that GHSR inverse agonism suppressed seizure severity in the rat amygdala kindling model, whereas GHSR antagonism made seizures more severe. Therefore, when considering the ghrelin system to modulate seizures, it is crucial to note the differential impact of various GHSR ligands.
Collapse
Affiliation(s)
- Siamak Beheshti
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Shiva Ershadi
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Fatemeh Zamani
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mansour Azimzadeh
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysis, 43400 UPM Serdang, Selangor, Malaysia
| | - Mohammad Wasil Wesal
- Department of Biology, Faculty of Education, University of Ghazni, Gazni, Afghanistan
| |
Collapse
|
27
|
Tuero C, Becerril S, Ezquerro S, Neira G, Frühbeck G, Rodríguez A. Molecular and cellular mechanisms underlying the hepatoprotective role of ghrelin against NAFLD progression. J Physiol Biochem 2023; 79:833-849. [PMID: 36417140 DOI: 10.1007/s13105-022-00933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022]
Abstract
The underlying mechanisms for the development and progression of nonalcoholic fatty liver disease (NAFLD) are complex and multifactorial. Within the last years, experimental and clinical evidences support the role of ghrelin in the development of NAFLD. Ghrelin is a gut hormone that plays a major role in the short-term regulation of appetite and long-term regulation of adiposity. The liver constitutes a target for ghrelin, where this gut-derived peptide triggers intracellular pathways regulating lipid metabolism, inflammation, and fibrosis. Interestingly, circulating ghrelin levels are altered in patients with metabolic diseases, such as obesity, type 2 diabetes, and metabolic syndrome, which, in turn, are well-known risk factors for the pathogenesis of NAFLD. This review summarizes the molecular and cellular mechanisms involved in the hepatoprotective action of ghrelin, including the reduction of hepatocyte lipotoxicity via autophagy and fatty acid β-oxidation, mitochondrial dysfunction, endoplasmic reticulum stress and programmed cell death, the reversibility of the proinflammatory phenotype in Kupffer cells, and the inactivation of hepatic stellate cells. Together, the metabolic and inflammatory pathways regulated by ghrelin in the liver support its potential as a therapeutic target to prevent NAFLD in patients with metabolic disorders.
Collapse
Affiliation(s)
- Carlota Tuero
- Department of General Surgery, Clínica Universidad de Navarra, School of Medicine, University of Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Silvia Ezquerro
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gabriela Neira
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain.
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
28
|
Bhargava R, Luur S, Rodriguez Flores M, Emini M, Prechtl CG, Goldstone AP. Postprandial Increases in Liver-Gut Hormone LEAP2 Correlate with Attenuated Eating Behavior in Adults Without Obesity. J Endocr Soc 2023; 7:bvad061. [PMID: 37287649 PMCID: PMC10243873 DOI: 10.1210/jendso/bvad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 06/09/2023] Open
Abstract
Background The novel liver-gut hormone liver-expressed antimicrobial peptide-2 (LEAP2) is a centrally acting inverse agonist, and competitive antagonist of orexigenic acyl ghrelin (AG), at the GH secretagogue receptor, reducing food intake in rodents. In humans, the effects of LEAP2 on eating behavior and mechanisms behind the postprandial increase in LEAP2 are unclear, though this is reciprocal to the postprandial decrease in plasma AG. Methods Plasma LEAP2 was measured in a secondary analysis of a previous study. Twenty-two adults without obesity attended after an overnight fast, consuming a 730-kcal meal without or with subcutaneous AG administration. Postprandial changes in plasma LEAP2 were correlated with postprandial changes in appetite, high-energy (HE) or low-energy (LE) food cue reactivity using functional magnetic resonance imaging, ad libitum food intake, and plasma/serum AG, glucose, insulin, and triglycerides. Results Postprandial plasma LEAP2 increased by 24.5% to 52.2% at 70 to 150 minutes, but was unchanged by exogenous AG administration. Postprandial increases in LEAP2 correlated positively with postprandial decreases in appetite, and cue reactivity to HE/LE and HE food in anteroposterior cingulate cortex, paracingulate cortex, frontal pole, and middle frontal gyrus, with similar trend for food intake. Postprandial increases in LEAP2 correlated negatively with body mass index, but did not correlate positively with increases in glucose, insulin, or triglycerides, nor decreases in AG. Conclusions These correlational findings are consistent with a role for postprandial increases in plasma LEAP2 in suppressing human eating behavior in adults without obesity. Postprandial increases in plasma LEAP2 are unrelated to changes in plasma AG and the mediator(s) remain uncertain.
Collapse
Affiliation(s)
- Raghav Bhargava
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Sandra Luur
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Marcela Rodriguez Flores
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Mimoza Emini
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Christina G Prechtl
- School of Public Health, Faculty of Medicine, Imperial College London, St. Mary's Hospital, London, W2 1PG, UK
| | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| |
Collapse
|
29
|
Tian J, Guo L, Wang T, Jia K, Swerdlow RH, Zigman JM, Du H. Liver-expressed antimicrobial peptide 2 elevation contributes to age-associated cognitive decline. JCI Insight 2023; 8:166175. [PMID: 37212281 DOI: 10.1172/jci.insight.166175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/31/2023] [Indexed: 05/23/2023] Open
Abstract
Elderly individuals frequently report cognitive decline, while various studies indicate hippocampal functional declines with advancing age. Hippocampal function is influenced by ghrelin through hippocampus-expressed growth hormone secretagogue receptor (GHSR). Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous GHSR antagonist that attenuates ghrelin signaling. Here, we measured plasma ghrelin and LEAP2 levels in a cohort of cognitively normal individuals older than 60 and found that LEAP2 increased with age while ghrelin (also referred to in literature as "acyl-ghrelin") marginally declined. In this cohort, plasma LEAP2/ghrelin molar ratios were inversely associated with Mini-Mental State Examination scores. Studies in mice showed an age-dependent inverse relationship between plasma LEAP2/ghrelin molar ratio and hippocampal lesions. In aged mice, restoration of the LEAP2/ghrelin balance to youth-associated levels with lentiviral shRNA Leap2 downregulation improved cognitive performance and mitigated various age-related hippocampal deficiencies such as CA1 region synaptic loss, declines in neurogenesis, and neuroinflammation. Our data collectively suggest that LEAP2/ghrelin molar ratio elevation may adversely affect hippocampal function and, consequently, cognitive performance; thus, it may serve as a biomarker of age-related cognitive decline. Moreover, targeting LEAP2 and ghrelin in a manner that lowers the plasma LEAP2/ghrelin molar ratio could benefit cognitive performance in elderly individuals for rejuvenation of memory.
Collapse
Affiliation(s)
- Jing Tian
- Department of Pharmacology and Toxicology and
| | - Lan Guo
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
| | - Tienju Wang
- Department of Pharmacology and Toxicology and
| | - Kun Jia
- Department of Pharmacology and Toxicology and
| | - Russell H Swerdlow
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffrey M Zigman
- Departments of Internal Medicine and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Heng Du
- Department of Pharmacology and Toxicology and
- Higuchi Biosciences Center, University of Kansas, Lawrence, Kansas, USA
- Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
30
|
Stark R, Feehan J, Mousa A, Andrews ZB, de Courten B. Liver-expressed antimicrobial peptide 2 is associated with improved pancreatic insulin secretion in adults with overweight and obesity. Diabetes Obes Metab 2023; 25:1213-1220. [PMID: 36597795 PMCID: PMC10947148 DOI: 10.1111/dom.14968] [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: 10/27/2022] [Revised: 12/15/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023]
Abstract
AIMS To examine association of liver-expressed antimicrobial peptide 2 (LEAP2), an endogenous ghrelin antagonist with anorexiant effects, to key cardiometabolic risk factors in people with overweight and obesity. METHODS In this cross-sectional study, we sought to identify associations between LEAP2 levels and cardiometabolic risk factors, including body composition (dual X-ray absorptiometry), insulin and glucose metabolism (oral and intravenous glucose tolerance tests and hyperinsulinaemic-euglycaemic clamps), plasma lipids and inflammation markers (ELISA and multiplex assays). RESULTS In 65 participants with overweight or obesity (63.1% male, mean age 31.3 ± 8.5 years), LEAP2 levels were associated with total body fat, but not with body mass index or waist-hip ratio in both univariable and age- and sex-adjusted models (P < 0.05). Higher LEAP2 level was also positively associated with higher insulin secretion in univariable (P = 0.047) and multivariable models adjusted for age, sex and body fat (P = 0.03), but not with fasting glucose levels (P ≥ 0.05). Higher LEAP2 levels were associated insulin resistance (P = 0.07) after adjustment for age and sex, but the association disappeared after an additional adjustment for body fat (P = 0.2). There was an inverse association between LEAP2 levels and nuclear factor kappa-B (NFκB) activity in the peripheral blood mononuclear cells in age-, sex- and body fat-adjusted models (P = 0.04). There were no associations with cardiovascular risk factors (lipids, blood pressure) or other inflammation markers. CONCLUSIONS These results provide important insights into the association between LEAP2 and cardiometabolic health in a high-risk population of individuals with overweight and obesity. This is a first report of an association between LEAP2 and insulin secretion, insulin sensitivity and NFκB activity. LEAP2 may represent an important potential therapeutic target to promote insulin secretion in people with type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Romana Stark
- School of Clinical SciencesMonash UniversityClaytonVictoriaAustralia
| | - Jack Feehan
- Institute for Health and Sport, Victoria UniversityFootscrayVictoriaAustralia
| | - Aya Mousa
- Monash Biomedicine Discovery Institute and Department of PhysiologyMonash UniversityClaytonVictoriaAustralia
| | - Zane B. Andrews
- School of Clinical SciencesMonash UniversityClaytonVictoriaAustralia
| | - Barbora de Courten
- Monash Centre for Health Research and Implementation (MCHRI), Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVictoriaAustralia
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
| |
Collapse
|
31
|
Nabekura H, Islam MN, Sakoda H, Yamaguchi T, Saiki A, Nabekura T, Oshiro T, Tanaka Y, Murayama S, Zhang W, Tatsuno I, Nakazato M. Liver-Expressed Antimicrobial Peptide 2 Is a Hepatokine That Predicts Weight Loss and Complete Remission of Type 2 Diabetes Mellitus after Vertical Sleeve Gastrectomy in Japanese Individuals. Obes Facts 2023; 16:392-400. [PMID: 37094564 PMCID: PMC10427959 DOI: 10.1159/000530733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
INTRODUCTION Vertical sleeve gastrectomy (VSG) is considered one of the most effective treatments for sustained weight loss and complete remission of type 2 diabetes mellitus (CR-T2DM). Liver-expressed antimicrobial peptide 2 (LEAP2), a ghrelin receptor antagonist peptide, is a metabolic hormone regulated by VSG. However, it is unknown whether LEAP2 can be used to predict the outcomes of VSG. This study aimed to evaluate LEAP2 as a predictive factor for weight loss and CR-T2DM after VSG. METHODS This retrospective study included 39 Japanese participants with obesity who underwent VSG. Serum LEAP2, des-acyl ghrelin (DAG), and other metabolic and anthropometric parameters were studied before and at 12 months after VSG. Receiver operating characteristics (ROC) curve was generated to evaluate predictive score for weight loss with cut-off value of >50 percent excess weight loss. ROC curve was also generated to assess CR-T2DM. RESULTS Serum LEAP2 levels were significantly higher in participants with body mass index (BMI) 32-50 kg/m2 than in those with normal weight. Participants with BMI >50 kg/m2 had lower serum LEAP2 concentrations than those with BMI 32-50 kg/m2. VSG caused a significant reduction in serum DAG concentrations, but it did not affect serum LEAP2 concentrations in either male or female participants. Preoperative serum LEAP2 concentration of 2.88 pmol/mL was the optimal cutoff value for predicting weight loss after VSG, with sensitivity of 80.0% and specificity of 75.9%. Preoperative serum LEAP2 level higher than 4.67 pmol/mL predicted CR-T2DM after VSG with sensitivity of 100% and specificity of 58.8%. CONCLUSION Preoperative serum LEAP2 could predict weight loss and CR-T2DM as outcomes of VSG.
Collapse
Affiliation(s)
- Hiroki Nabekura
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Md Nurul Islam
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hideyuki Sakoda
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan,
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan,
| | - Takashi Yamaguchi
- Center of Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, Chiba, Japan
| | - Atsuhito Saiki
- Center of Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, Chiba, Japan
| | - Taiki Nabekura
- Department of Surgery, Toho University Sakura Medical Center, Chiba, Japan
| | - Takashi Oshiro
- Department of Surgery, Toho University Sakura Medical Center, Chiba, Japan
| | - Yuri Tanaka
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shinya Murayama
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Weidong Zhang
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Ichiro Tatsuno
- Center of Diabetes, Endocrinology and Metabolism, Toho University Sakura Medical Center, Chiba, Japan
- Chiba Prefectural University of Health Sciences, Chiba, Japan
| | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Inter-Organ Communication Research Project, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
- AMED-CREST, Agency for Medical Research and Development, Tokyo, Japan
- Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
32
|
Wald HS, Ghidewon MY, Hayes MR, Grill HJ. Hindbrain ghrelin and liver-expressed antimicrobial peptide 2, ligands for growth hormone secretagogue receptor, bidirectionally control food intake. Am J Physiol Regul Integr Comp Physiol 2023; 324:R547-R555. [PMID: 36847494 PMCID: PMC10069974 DOI: 10.1152/ajpregu.00232.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 03/01/2023]
Abstract
Hindbrain growth hormone secretagogue receptor (GHSR) agonism increases food intake, yet the underlying neural mechanisms remain unclear. The functional effects of hindbrain GHSR antagonism by its endogenous antagonist liver-expressed antimicrobial peptide 2 (LEAP2) are also yet unexplored. To test the hypothesis that hindbrain GHSR agonism attenuates the food intake inhibitory effect of gastrointestinal (GI) satiation signals, ghrelin (at a feeding subthreshold dose) was administered to the fourth ventricle (4V) or directly to the nucleus tractus solitarius (NTS) before systemic delivery of the GI satiation signal cholecystokinin (CCK). Also examined, was whether hindbrain GHSR agonism attenuated CCK-induced NTS neural activation (c-Fos immunofluorescence). To investigate an alternate hypothesis that hindbrain GHSR agonism enhances feeding motivation and food seeking, intake stimulatory ghrelin doses were administered to the 4V and fixed ratio 5 (FR-5), progressive ratio (PR), and operant reinstatement paradigms for palatable food responding were evaluated. Also assessed were 4V LEAP2 delivery on food intake and body weight (BW) and on ghrelin-stimulated feeding. Both 4V and NTS ghrelin blocked the intake inhibitory effect of CCK and 4V ghrelin blocked CCK-induced NTS neural activation. Although 4V ghrelin increased low-demand FR-5 responding, it did not increase high-demand PR or reinstatement of operant responding. Fourth ventricle LEAP2 reduced chow intake and BW and blocked hindbrain ghrelin-stimulated feeding. Data support a role for hindbrain GHSR in bidirectional control of food intake through mechanisms that include interacting with the NTS neural processing of GI satiation signals but not food motivation and food seeking.
Collapse
Affiliation(s)
- Hallie S Wald
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Misgana Y Ghidewon
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Matthew R Hayes
- Department of Psychiatry, Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Harvey J Grill
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
33
|
Uchida Y, Izumizaki M. Modulation of acyl and des-acyl ghrelin on autonomic and behavioral thermoregulation in various ambient temperatures. J Therm Biol 2023; 113:103543. [PMID: 37055119 DOI: 10.1016/j.jtherbio.2023.103543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/13/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023]
Abstract
Maintenance of body temperature (Tb) at various ambient temperatures (Ta) during fasting is important for homeotherms. Fasting decreases Tb in thermoneutral and cold conditions and facilitates thermoregulatory behavior in the cold in rats; however, the mechanism is unknown. We focused on ghrelin, a hormone secreted by the stomach during fasting, in two circulatory forms: acyl ghrelin (AG) and des-acyl ghrelin (DAG). AG is called active ghrelin, while DAG, the non-active ghrelin, was unknown for a long time before its many functions were recently clarified. In the present review, we present the modulation of AG and DAG on autonomic and behavioral thermoregulation at various Ta and discuss the differences between their modulation on thermoregulation. AG decreases Tb in thermoneutral and cold conditions but does not affect the thermoregulatory behavior of rodents in cold conditions. The DAG decreases Tb in thermoneutral and hot conditions, but it does not affect Tb and facilitates the thermoregulatory behavior of rodents in the cold. These findings indicate that the actions of AG and DAG on thermoregulation are similar in thermoneutral conditions but are different in cold conditions.
Collapse
|
34
|
Tezenas-du-Montcel C, Tolle V. La régulation de la prise alimentaire au travers des actions antagonistes de la ghréline et du LEAP-2. CAHIERS DE NUTRITION ET DE DIÉTÉTIQUE 2023. [DOI: 10.1016/j.cnd.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
35
|
Gross JD, Zhou Y, Barak LS, Caron MG. Ghrelin receptor signaling in health and disease: a biased view. Trends Endocrinol Metab 2023; 34:106-118. [PMID: 36567228 PMCID: PMC9852078 DOI: 10.1016/j.tem.2022.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 12/06/2022] [Indexed: 12/25/2022]
Abstract
As allosteric complexes, G-protein-coupled receptors (GPCRs) respond to extracellular stimuli and pleiotropically couple to intracellular transducers to elicit signaling pathway-dependent effects in a process known as biased signaling or functional selectivity. One such GPCR, the ghrelin receptor (GHSR1a), has a crucial role in restoring and maintaining metabolic homeostasis during disrupted energy balance. Thus, pharmacological modulation of GHSR1a bias could offer a promising strategy to treat several metabolism-based disorders. Here, we summarize current evidence supporting GHSR1a functional selectivity in vivo and highlight recent structural data. We propose that precise determinations of GHSR1a molecular pharmacology and pathway-specific physiological effects will enable discovery of GHSR1a drugs with tailored signaling profiles, thereby providing safer and more effective treatments for metabolic diseases.
Collapse
Affiliation(s)
- Joshua D Gross
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Yang Zhou
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Lawrence S Barak
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| | - Marc G Caron
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
36
|
Ragland TJ, Malin SK. Plasma LEAP-2 Following a Low-Calorie Diet with or without Interval Exercise in Women with Obesity. Nutrients 2023; 15:655. [PMID: 36771362 PMCID: PMC9918887 DOI: 10.3390/nu15030655] [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: 12/06/2022] [Revised: 01/14/2023] [Accepted: 01/22/2023] [Indexed: 02/01/2023] Open
Abstract
Liver-expressed antimicrobial peptide-2 (LEAP-2) is associated with caloric intake and glucose metabolism. Purpose: Assess if a low-calorie diet with interval exercise (LCD+INT) raises LEAP-2 more than LCD in relation to appetite and cardiometabolic health. Methods: Women with obesity were randomized to either 2 weeks of LCD (n = 13, ~1200 kcal/d) or LCD+INT (n = 12; 60 min/d) of INT at 3 min of 90% and 50% HRpeak, respectively. LEAP-2 and acylated ghrelin (AG) were measured at 0, 30, and 60 min, while glucose, insulin, C-peptide, and free fatty acids (FFA) were obtained up to 180 min of a 75 g OGTT. Fasting and 120 min OGTT appetite were assessed via visual analog scales. Results: LCD reduced the BMI (p < 0.001) compared with LCD+INT, but only LCD+INT increased the VO2 max (p = 0.04). Treatments reduced fasting LEAP-2 (p = 0.05), but only LCD increased LEAP-2 iAUC60 min (p = 0.06) and post-prandial LEAP-2 stimulation (p = 0.02). Higher post-LEAP-260 min tended to relate to a lower desire to eat 120 min of sweet (r = 0.40, p = 0.07) and salty foods (r = 0.41, p = 0.06), as well as lower AG30 min (r = -0.51, p = 0.01) and higher FFA iAUC180 min (r = 0.56, p = 0.007) post-treatment. Conclusion: LCD, with or without INT, reduced fasting LEAP-2, but only LCD raised post-prandial LEAP-2. How diet and exercise impact LEAP-2 for lower chronic disease risk awaits further investigation.
Collapse
Affiliation(s)
| | - Steven K. Malin
- Department of Kinesiology & Health, New Brunswick, NJ 08091, USA
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers University, New Brunswick, NJ 08091, USA
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ 08091, USA
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, NJ 08091, USA
| |
Collapse
|
37
|
Wada R, Takemi S, Matsumoto M, Iijima M, Sakai T, Sakata I. Molecular cloning and analysis of the ghrelin/GHSR system in Xenopus tropicalis. Gen Comp Endocrinol 2023; 331:114167. [PMID: 36402245 DOI: 10.1016/j.ygcen.2022.114167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/16/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
Abstract
Ghrelin is a gut-derived peptide with several physiological functions, including feeding, gastrointestinal motility, and hormonal secretion. Recently, a host defense peptide, liver-expressed antimicrobial peptide-2 (LEAP2), was reported as an endogenous antagonist of growth hormone secretagogue receptor (GHS-R). The physiological relevance of the molecular LEAP2-GHS-R interaction in mammals has been explored; however, studies on non-mammals are limited. Here, we report the identification and functional characterization of ghrelin and its related molecules in Western clawed frog (Xenopus tropicalis), a known model organism. We first identified cDNA encoding X. tropicalis ghrelin and GHS-R. RT-qPCR revealed that ghrelin mRNA expression was most abundant in the stomach. GHS-R mRNA was widely distributed in the brain and peripheral tissues, and a relatively strong signal was observed in the stomach and intestine. In addition, LEAP2 was mainly expressed in intestinal tissues at higher levels than in the liver. In functional analysis, X. tropicalis ghrelin and human ghrelin induced intracellular Ca2+ mobilization with EC50 values in the low nanomolar range in CHO-K1 cells expressing X. tropicalis GHS-R. Furthermore, ghrelin-induced GHS-R activation was antagonized with IC50 values in the nanomolar range by heterologous human LEAP2. We also validated the expression of ghrelin and feeding-related factors under fasting conditions. After 2 days of fasting, no changes in ghrelin mRNA levels were observed in the stomach, but GHS-R mRNA levels were significantly increased, associated with significant downregulation of nucb2. In addition, LEAP2 upregulation was observed in the duodenum. These results provide the first evidence that LEAP2 functions as an antagonist of GHS-R in the anuran amphibian X. tropicalis. It has also been suggested that the ghrelin/GHS-R/LEAP2 system may be involved in energy homeostasis in X. tropicalis.
Collapse
Affiliation(s)
- Reiko Wada
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama 338-8570, Japan
| | - Shota Takemi
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama 338-8570, Japan
| | - Mio Matsumoto
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama 338-8570, Japan
| | - Mio Iijima
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama 338-8570, Japan
| | - Takafumi Sakai
- Saitama University, 255 Shimo-okubo, Sakura-ku, Saitama 338-8570, Japan
| | - Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-ohkubo, Sakuraku, Saitama 338-8570, Japan.
| |
Collapse
|
38
|
Jerlhag E. Animal studies reveal that the ghrelin pathway regulates alcohol-mediated responses. Front Psychiatry 2023; 14:1050973. [PMID: 36970276 PMCID: PMC10030715 DOI: 10.3389/fpsyt.2023.1050973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
Alcohol use disorder (AUD) is often described as repeated phases of binge drinking, compulsive alcohol-taking, craving for alcohol during withdrawal, and drinking with an aim to a reduce the negative consequences. Although multifaceted, alcohol-induced reward is one aspect influencing the former three of these. The neurobiological mechanisms regulating AUD processes are complex and one of these systems is the gut-brain peptide ghrelin. The vast physiological properties of ghrelin are mediated via growth hormone secretagogue receptor (GHSR, ghrelin receptor). Ghrelin is well known for its ability to control feeding, hunger, and metabolism. Moreover, ghrelin signaling appears central for alcohol-mediated responses; findings reviewed herein. In male rodents GHSR antagonism reduces alcohol consumption, prevents relapse drinking, and attenuates the motivation to consume alcohol. On the other hand, ghrelin increases the consumption of alcohol. This ghrelin-alcohol interaction is also verified to some extent in humans with high alcohol consumption. In addition, either pharmacological or genetic suppression of GHSR decreases several alcohol-related effects (behavioral or neurochemical). Indeed, this suppression blocks the alcohol-induced hyperlocomotion and dopamine release in nucleus accumbens as well as ablates the alcohol reward in the conditioned place preference model. Although not fully elucidated, this interaction appears to involve areas central for reward, such as the ventral tegmental area (VTA) and brain nodes targeted by VTA projections. As reviewed briefly, the ghrelin pathway does not only modulate alcohol-mediated effects, it regulates reward-related behaviors induced by addictive drugs. Although personality traits like impulsivity and risk-taking behaviors are common in patients with AUD, the role of the ghrelin pathway thereof is unknown and remains to be studied. In summary, the ghrelin pathway regulates addiction processes like AUD and therefore the possibility that GHSR antagonism reduces alcohol or drug-taking should be explored in randomized clinical trials.
Collapse
|
39
|
Chen RB, Wang QY, Wang YY, Wang YD, Liu JH, Liao ZZ, Xiao XH. Feeding-induced hepatokines and crosstalk with multi-organ: A novel therapeutic target for Type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1094458. [PMID: 36936164 PMCID: PMC10020511 DOI: 10.3389/fendo.2023.1094458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Hyperglycemia, which can be caused by either an insulin deficit and/or insulin resistance, is the main symptom of Type 2 diabetes, a significant endocrine metabolic illness. Conventional medications, including insulin and oral antidiabetic medicines, can alleviate the signs of diabetes but cannot restore insulin release in a physiologically normal amount. The liver detects and reacts to shifts in the nutritional condition that occur under a wide variety of metabolic situations, making it an essential organ for maintaining energy homeostasis. It also performs a crucial function in glucolipid metabolism through the secretion of hepatokines. Emerging research shows that feeding induces hepatokines release, which regulates glucose and lipid metabolism. Notably, these feeding-induced hepatokines act on multiple organs to regulate glucolipotoxicity and thus influence the development of T2DM. In this review, we focus on describing how feeding-induced cross-talk between hepatokines, including Adropin, Manf, Leap2 and Pcsk9, and metabolic organs (e.g.brain, heart, pancreas, and adipose tissue) affects metabolic disorders, thus revealing a novel approach for both controlling and managing of Type 2 diabetes as a promising medication.
Collapse
Affiliation(s)
- Rong-Bin Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qi-Yu Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiang-Hua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe-Zhen Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- *Correspondence: Xin-Hua Xiao, ; Zhe-Zhen Liao,
| | - Xin-Hua Xiao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- *Correspondence: Xin-Hua Xiao, ; Zhe-Zhen Liao,
| |
Collapse
|
40
|
Chicken LEAP2 Level Substantially Changes with Feed Intake and May Be Regulated by CDX4 in Small Intestine. Animals (Basel) 2022; 12:ani12243496. [PMID: 36552416 PMCID: PMC9774203 DOI: 10.3390/ani12243496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Ghrelin O-acyltransferase (GOAT), ghrelin, and GHSR have been reported to play important roles that influence feed intake in mammals. LEAP2, an endogenous antagonist of GHSR, plays an important role in the regulation of feed intake. However, chicken ghrelin has also been reported to have an inhibitory effect on feed intake. The role of the GOAT-Ghrelin-GHSR-LEAP2 axis in chicken-feed intake remains unclear. Therefore, it is necessary to systematically evaluate the changes in the tissue expression levels of these genes under different energy states. In this study, broiler chicks in different energy states were subjected to starvation and feeding, and relevant gene expression levels were measured using quantitative real-time PCR. Different energy states significantly modulated the expression levels of LEAP2 and GHSR but did not significantly affect the expression levels of GOAT and ghrelin. A high expression level of LEAP2 was detected in the liver and the whole small intestine. Compared to the fed group, the fasted chicks showed significantly reduced LEAP2 expression levels in the liver and the small intestine; 2 h after being refed, the LEAP2 expression of the fasted chicks returned to the level of the fed group. Transcription factor prediction and results of a dual luciferase assay indicated that the transcription factor CDX4 binds to the LEAP2 promoter region and positively regulates its expression. High expression levels of GHSR were detected in the hypothalamus and pituitary. Moreover, we detected GHSR highly expressed in the jejunum-this finding has not been previously reported. Thus, GHSR may regulate intestinal motility, and this aspect needs further investigation. In conclusion, this study revealed the function of chicken LEAP2 as a potential feed-intake regulator and identified the potential mechanism governing its intestine-specific expression. Our study lays the foundations for future studies on avian feed-intake regulation.
Collapse
|
41
|
Kharbanda KK, Farokhnia M, Deschaine SL, Bhargava R, Rodriguez-Flores M, Casey CA, Goldstone AP, Jerlhag E, Leggio L, Rasineni K. Role of the ghrelin system in alcohol use disorder and alcohol-associated liver disease: A narrative review. Alcohol Clin Exp Res 2022; 46:2149-2159. [PMID: 36316764 PMCID: PMC9772086 DOI: 10.1111/acer.14967] [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: 08/19/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 11/07/2022]
Abstract
Unhealthy alcohol consumption is a global health problem. Adverse individual, public health, and socioeconomic consequences are attributable to harmful alcohol use. Epidemiological studies have shown that alcohol use disorder (AUD) and alcohol-associated liver disease (ALD) are the top two pathologies among alcohol-related diseases. Consistent with the major role that the liver plays in alcohol metabolism, uncontrolled drinking may cause significant damage to the liver. This damage is initiated by excessive fat accumulation in the liver, which can further progress to advanced liver disease. The only effective therapeutic strategies currently available for ALD are alcohol abstinence or liver transplantation. Any molecule with dual-pronged effects at the central and peripheral organs controlling addictive behaviors and associated metabolic pathways are a potentially important therapeutic target for treating AUD and ALD. Ghrelin, a hormone primarily derived from the stomach, has such properties, and regulates both behavioral and metabolic functions. In this review, we highlight recent advances in understanding the peripheral and central functions of the ghrelin system and its role in AUD and ALD pathogenesis. We first discuss the correlation between blood ghrelin concentrations and alcohol use or abstinence. Next, we discuss the role of ghrelin in alcohol-seeking behaviors and finally its role in the development of fatty liver by metabolic regulations and organ crosstalk. We propose that a better understanding of the ghrelin system could open an innovative avenue for improved treatments for AUD and associated medical consequences, including ALD.
Collapse
Affiliation(s)
- Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland, USA
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sara L. Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland, USA
| | - Raghav Bhargava
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Marcela Rodriguez-Flores
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Carol A. Casey
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anthony P. Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland, USA
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island, USA
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
42
|
Holá L, Železná B, Karnošová A, Kuneš J, Fehrentz JA, Denoyelle S, Cantel S, Blechová M, Sýkora D, Myšková A, Maletínská L. A Novel Truncated Liver Enriched Antimicrobial Peptide-2 Palmitoylated at its N-Terminal Antagonizes Effects of Ghrelin. J Pharmacol Exp Ther 2022; 383:129-136. [PMID: 36198495 DOI: 10.1124/jpet.122.001322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/03/2022] [Indexed: 01/07/2023] Open
Abstract
Ghrelin is secreted in the stomach during fasting and targets the growth hormone secretagogue receptor (GHSR1a) in the hypothalamus and brainstem to exert its orexigenic effect. Recently, liver enriched antimicrobial peptide-2 (LEAP2) was identified as an endogenous high-affinity GHSR1a antagonist. LEAP2 is a 40-amino acid peptide with two disulfide bridges and GHRS1a affinity in the N-terminal hydrophobic part. In this study, we tested modified truncated N-terminal peptide LEAP2 (1-14), along with its myristoylated, palmitoylated, and stearoylated analogs, to determine their affinity to and activation of GHSR1a and their anorexigenic effects after acute peripheral administration. The lipidized analogs bound GHSR1a with affinity similar to that of natural LEAP2, and lipidization significantly enhanced the affinity of LEAP2(1-14) to GHSR1a. According to the beta-lactamase reporter gene response, the natural GHSR1a agonist ghrelin activated the receptor with nanomolar EC50 LEAP2(1-14) analogs behaved as inverse agonists of GHSR1a and suppressed internal activity of the receptor with EC50 values in the 10-8 M range. LEAP2(1-14) analogs significantly lowered acute food intake in overnight fasted mice, and palmitoylated LEAP2(1-14) was the most potent. In free-fed mice, all LEAP2(1-14) analogs significantly decreased the orexigenic effect of the stable ghrelin analog [Dpr3]Ghrelin. Moreover, palmitoylated LEAP2(1-14) inhibited the growth hormone (GH) release induced by [Dpr3] Ghrelin and exhibited an increased stability in rat plasma compared with LEAP2(1-14). In conclusion, palmitoylated LEAP2(1-14) had the most pronounced affinity for GHSR1a, had an anorexigenic effect, exhibited stability in rat plasma, and attenuated [Dpr3]Ghrelin-induced GH release. Such properties render palmitoylated LEAP2(1-14) a promising substance for antiobesity treatment. SIGNIFICANCE STATEMENT: The agonist and antagonist of one receptor are rarely found in one organism. For ghrelin receptor (growth hormone secretagogue receptor, GHSR), endogenous agonist ghrelin and endogenous antagonist/inverse agonist liver enriched antimicrobial peptide-2 (LEAP2) co-exist and differently control GHSR signaling. As ghrelin has a unique role in food intake regulation, energy homeostasis, and cytoprotection, lipidized truncated LEAP2 analogs presented in this study could serve not only to reveal the relationship between ghrelin and LEAP2 but also for development of potential anti-obesity agents.
Collapse
Affiliation(s)
- Lucie Holá
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Alena Karnošová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Jean-Alain Fehrentz
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Séverine Denoyelle
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Sonia Cantel
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Miroslava Blechová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - David Sýkora
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Aneta Myšková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic (L.H., B.Ž., A.K., J.K., M.B., A.M., L.M.); Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (J.K.); First Faculty of Medicine, Charles University, Prague, Czech Republic (L.H., A.K.); University of Chemistry and Technology, Prague, Czech Republic (D.S., A.M.); and IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France (J.A.F., S.C., S.D.)
| |
Collapse
|
43
|
Regulation of feeding and therapeutic application of bioactive peptides. Pharmacol Ther 2022; 239:108187. [DOI: 10.1016/j.pharmthera.2022.108187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
44
|
Li HZ, Shao XX, Shou LL, Li N, Liu YL, Xu ZG, Guo ZY. Development of Esterase-Resistant and Highly Active Ghrelin Analogs via Thiol-Ene Click Chemistry. ACS Med Chem Lett 2022; 13:1655-1662. [PMID: 36262400 PMCID: PMC9575166 DOI: 10.1021/acsmedchemlett.2c00339] [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: 07/22/2022] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
The orexigenic peptide ghrelin exerts important functions in energy metabolism and has therapeutic potential to treat certain diseases. Native ghrelin carries an essential O-fatty acyl moiety; however, this post-translational modification is susceptible to hydrolysis by certain esterases in circulation, representing a major route of its in vivo inactivation. In the present study, we developed a novel approach to prepare various esterase-resistant ghrelin analogs via photoinduced thiol-ene click chemistry. A recombinant unacylated human ghrelin mutant was reacted with commercially available terminal alkenes; thus, various alkyl moieties were introduced to the side chain of its unique Cys3 residue via a thioether bond. Among 11 S-alkylated ghrelin analogs, analog 11, generated by reacting with 2-methyl-1-octene, not only acquired much higher stability in serum but also retained full activity compared with native human ghrelin. Thus, the present study provided an efficient approach to prepare highly stable and highly active ghrelin analogs with therapeutic potential.
Collapse
Affiliation(s)
- Hao-Zheng Li
- Research Center for Translational Medicine
at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiao-Xia Shao
- Research Center for Translational Medicine
at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Li-Li Shou
- Research Center for Translational Medicine
at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ning Li
- Research Center for Translational Medicine
at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ya-Li Liu
- Research Center for Translational Medicine
at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zeng-Guang Xu
- Research Center for Translational Medicine
at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zhan-Yun Guo
- Research Center for Translational Medicine
at East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| |
Collapse
|
45
|
Conde K, Kulyk D, Vanschaik A, Daisey S, Rojas C, Wiersielis K, Yasrebi A, Degroat TJ, Sun Y, Roepke TA. Deletion of Growth Hormone Secretagogue Receptor in Kisspeptin Neurons in Female Mice Blocks Diet-Induced Obesity. Biomolecules 2022; 12:1370. [PMID: 36291579 PMCID: PMC9599822 DOI: 10.3390/biom12101370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 01/19/2023] Open
Abstract
The gut peptide, ghrelin, mediates energy homeostasis and reproduction by acting through its receptor, growth hormone secretagogue receptor (GHSR), expressed in hypothalamic neurons in the arcuate (ARC). We have shown 17β-estradiol (E2) increases Ghsr expression in Kisspeptin/Neurokinin B/Dynorphin (KNDy) neurons, enhancing sensitivity to ghrelin. We hypothesized that E2-induced Ghsr expression augments KNDy sensitivity in a fasting state by elevating ghrelin to disrupt energy expenditure in females. We produced a Kiss1-GHSR knockout to determine the role of GHSR in ARC KNDy neurons. We found that changes in ARC gene expression with estradiol benzoate (EB) treatment were abrogated by the deletion of GHSR and ghrelin abolished these differences. We also observed changes in metabolism and fasting glucose levels. Additionally, knockouts were resistant to body weight gain on a high fat diet (HFD). Behaviorally, we found that knockouts on HFD exhibited reduced anxiety-like behavior. Furthermore, knockouts did not refeed to the same extent as controls after a 24 h fast. Finally, in response to cold stress, knockout females had elevated metabolic parameters compared to controls. These data indicate GHSR in Kiss1 neurons modulate ARC gene expression, metabolism, glucose homeostasis, behavior, and thermoregulation, illustrating a novel mechanism for E2 and ghrelin to control Kiss1 neurons.
Collapse
Affiliation(s)
- Kristie Conde
- Graduate Program in Neuroscience, Rutgers University Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Danielle Kulyk
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Allison Vanschaik
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Sierra Daisey
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Catherine Rojas
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Kimberly Wiersielis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Thomas J. Degroat
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Troy A. Roepke
- Graduate Program in Neuroscience, Rutgers University Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Rutgers Center for Lipid Research, the Center for Nutrition, Microbiome, and Health, and the New Jersey Institute of Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
46
|
Study of the Ghrelin/LEAP-2 Ratio in Humans and Rats during Different Phases of Pregnancy. Int J Mol Sci 2022; 23:ijms23179514. [PMID: 36076912 PMCID: PMC9455743 DOI: 10.3390/ijms23179514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 12/04/2022] Open
Abstract
The Liver-Expressed Antimicrobial Peptide 2 (LEAP-2) has emerged as an endogenous GHS-R antagonist and blunts the orexigenic action of ghrelin. This study aimed to determine the Ghrelin/LEAP-2 ratio in humans and rats during pregnancy. In humans, we conducted a nested case-control study within an observational prospective cohort. Healthy and mild preeclamptic pregnant women were studied at each trimester of gestation and three months postpartum. In addition, a group of non-pregnant women was studied into the follicular and luteal phases of the menstrual cycle. Furthermore, Ghrelin/LEAP-2 ratio was investigated in non-pregnant rats and at different periods of rat pregnancy. Human and rat serum ghrelin and LEAP-2 levels were determined using the commercially available ELISA kits. The Ghrelin/LEAP-2 ratio peak around the second trimester of gestation in healthy pregnant women (p < 0.05). Additionally, there were no statistically significant differences in Ghrelin/LEAP-2 ratio between healthy and preeclamptic pregnant women at each trimester of gestation (p > 0.05). The Ghrelin/LEAP-2 ratio in pregnant rat reached the peak around mid-gestation with a similar pattern to the human pregnancy. LEAP-2 was visualized by immunohistochemistry in human term placenta and rat placentas on days 12, 16 and 21 of pregnancy. In conclusion, this study provides the first evidence of a Ghrelin/LEAP-2 ratio peak around the half-way point of pregnancy onwards during human and rat pregnancy, and it might be associated with increased rates of weight gain during pregnancy. Thus, this study suggests that LEAP-2 and Ghrelin/LEAP-2 ratio might play an important role in maternal physiology adaptation of weight gain during pregnancy.
Collapse
|
47
|
Involvement of the ghrelin system in the maintenance and reinstatement of cocaine-motivated behaviors: a role of adrenergic action at peripheral β1 receptors. Neuropsychopharmacology 2022; 47:1449-1460. [PMID: 34923576 PMCID: PMC9206024 DOI: 10.1038/s41386-021-01249-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/30/2022]
Abstract
Cocaine addiction is a significant medical and public concern. Despite decades of research effort, development of pharmacotherapy for cocaine use disorder remains largely unsuccessful. This may be partially due to insufficient understanding of the complex biological mechanisms involved in the pathophysiology of this disorder. In the present study, we show that: (1) elevation of ghrelin by cocaine plays a critical role in maintenance of cocaine self-administration and cocaine-seeking motivated by cocaine-conditioned stimuli; (2) acquisition of cocaine-taking behavior is associated with the acquisition of stimulatory effects of cocaine by cocaine-conditioned stimuli on ghrelin secretion, and with an upregulation of ghrelin receptor mRNA levels in the ventral tegmental area (VTA); (3) blockade of ghrelin signaling by pretreatment with JMV2959, a selective ghrelin receptor antagonist, dose-dependently inhibits reinstatement of cocaine-seeking triggered by either cocaine or yohimbine in behaviorally extinguished animals with a history of cocaine self-administration; (4) JMV2959 pretreatment also inhibits brain stimulation reward (BSR) and cocaine-potentiated BSR maintained by optogenetic stimulation of VTA dopamine neurons in DAT-Cre mice; (5) blockade of peripheral adrenergic β1 receptors by atenolol potently attenuates the elevation in circulating ghrelin induced by cocaine and inhibits cocaine self-administration and cocaine reinstatement triggered by cocaine. These findings demonstrate that the endogenous ghrelin system plays an important role in cocaine-related addictive behaviors and suggest that manipulating and targeting this system may be viable for mitigating cocaine use disorder.
Collapse
|
48
|
Holm S, Husted AS, Skov LJ, Morville TH, Hagemann CA, Jorsal T, Dall M, Jakobsen A, Klein AB, Treebak JT, Knop FK, Schwartz TW, Clemmensen C, Holst B. Beta-Hydroxybutyrate Suppresses Hepatic Production of the Ghrelin Receptor Antagonist LEAP2. Endocrinology 2022; 163:6555773. [PMID: 35352108 PMCID: PMC9119693 DOI: 10.1210/endocr/bqac038] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Liver-expressed antimicrobial peptide-2 (LEAP2) is an endogenous ghrelin receptor antagonist, which is upregulated in the fed state and downregulated during fasting. We hypothesized that the ketone body beta-hydroxybutyrate (BHB) is involved in the downregulation of LEAP2 during conditions with high circulating levels of BHB. METHODS Hepatic and intestinal Leap2 expression were determined in 3 groups of mice with increasing circulating levels of BHB: prolonged fasting, prolonged ketogenic diet, and oral BHB treatment. LEAP2 levels were measured in lean and obese individuals, in human individuals following endurance exercise, and in mice after BHB treatment. Lastly, we investigated Leap2 expression in isolated murine hepatocytes challenged with BHB. RESULTS We confirmed increased circulating LEAP2 levels in individuals with obesity compared to lean individuals. The recovery period after endurance exercise was associated with increased plasma levels of BHB levels and decreased LEAP2 levels in humans. Leap2 expression was selectively decreased in the liver after fasting and after exposure to a ketogenic diet for 3 weeks. Importantly, we found that oral administration of BHB increased circulating levels of BHB in mice and decreased Leap2 expression levels and circulating LEAP2 plasma levels, as did Leap2 expression after direct exposure to BHB in isolated murine hepatocytes. CONCLUSION From our data, we suggest that LEAP2 is downregulated during different states of energy deprivation in both humans and rodents. Furthermore, we here provide evidence that the ketone body, BHB, which is highly upregulated during fasting metabolism, directly downregulates LEAP2 levels. This may be relevant in ghrelin receptor-induced hunger signaling during energy deprivation.
Collapse
Affiliation(s)
- Stephanie Holm
- Correspondence: Stephanie Holm, MSc, Novo Nordisk Foundation Center for Basic Metabolic Research (6th floor) & Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| | - Anna S Husted
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Louise J Skov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thomas H Morville
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christoffer A Hagemann
- Center for Clinical Metabolic Research, Copenhagen University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark
- Gubra, 2970 Hørsholm, Denmark
| | - Tina Jorsal
- Center for Clinical Metabolic Research, Copenhagen University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark
| | - Morten Dall
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alexander Jakobsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Anders B Klein
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Filip K Knop
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Clinical Metabolic Research, Copenhagen University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Birgitte Holst
- Correspondence: Birgitte Holst MD, PhD, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
49
|
Islam MN, Zhang W, Sakai K, Nakazato Y, Tanida R, Sakoda H, Takei T, Takao T, Nakazato M. Liver-expressed antimicrobial peptide 2 functions independently of growth hormone secretagogue receptor in calorie-restricted mice. Peptides 2022; 151:170763. [PMID: 35151767 DOI: 10.1016/j.peptides.2022.170763] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022]
Abstract
Ghrelin is a gastric-derived peptide that stimulates feeding, blood glucose elevation, body temperature reduction, and growth hormone (GH) secretion. Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous antagonist of the ghrelin receptor, also called growth hormone secretagogue receptor (GHSR). We studied the effects of LEAP2 administration on feeding, body weight, glycemia, body temperature, and inflammation-related genes in the liver in C57BL/6 J mice and Ghsr-knockout (Ghsr-KO) mice. We found that a single administration of LEAP2 did not abolish fasting-induced food intake in 24-h fasted C57BL/6 J mice or Ghsr-KO mice. Moreover, continuous LEAP2 administration to mice fed ad libitum for 6 days did not affect feeding, body temperature, plasma ghrelin, or blood glucose. By contrast, continuous LEAP2 administration to calorie-restricted C57BL/6 J mice and Ghsr-KO mice induced body weight loss, hypoglycemia, body temperature reduction, and upregulation of Il-6 and Il-1β mRNAs in the liver. Our findings suggest that LEAP2 functions independently of GHSR, implying that LEAP2 affects physiology beyond the ghrelin-GHSR system.
Collapse
Affiliation(s)
- Md Nurul Islam
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Weidong Zhang
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Katsuya Sakai
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuki Nakazato
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Ryota Tanida
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Ishikawa 920-8640, Japan
| | - Hideyuki Sakoda
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Toshiki Takei
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Toshifumi Takao
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Masamitsu Nakazato
- Division of Interactive Organ Systems, Department of Projects Research, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan; AMED-CREST, Agency for Medical Research and Development, Tokyo 100-0004, Japan.
| |
Collapse
|
50
|
Péraldi-Roux S, Bayle M, M'Kadmi C, Damian M, Vaillé J, Fernandez G, Paula Cornejo M, Marie J, Banères JL, Ben Haj Salah K, Fehrentz JA, Cantel S, Perello M, Denoyelle S, Oiry C, Neasta J. Design and Characterization of a Triazole-Based Growth Hormone Secretagogue Receptor Modulator Inhibiting the Glucoregulatory and Feeding Actions of Ghrelin. Biochem Pharmacol 2022; 202:115114. [DOI: 10.1016/j.bcp.2022.115114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 11/02/2022]
|