1
|
Johansen VBI, Gradel AKJ, Holm SK, Cuenco J, Merrild C, Petersen N, Demozay D, Mani BK, Suppli MP, Grøndahl MFG, Lund AB, Knop FK, Prada-Medina CA, Hogendorf WFJ, Lykkesfeldt J, Merkestein M, Sakamoto K, Holst B, Clemmensen C. Regulation of LEAP2 by insulin and glucagon in mice and humans. Cell Rep Med 2025; 6:101996. [PMID: 40056903 DOI: 10.1016/j.xcrm.2025.101996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 09/14/2024] [Accepted: 02/10/2025] [Indexed: 03/10/2025]
Abstract
Liver-expressed antimicrobial peptide 2 (LEAP2) is an endogenous antagonist and inverse agonist of the ghrelin receptor, countering ghrelin's effects on cell signaling and feeding. However, despite an emerging interest in LEAP2's physiology and pharmacology, its endocrine regulation remains unclear. Here, we report that plasma LEAP2 levels decrease significantly upon glucagon infusions during somatostatin clamps in humans. This effect is preserved in patients with obesity and type 2 diabetes while diminished following a hypercaloric diet and a sedentary lifestyle for 2 weeks. Additionally, insulin receptor antagonism offsets the upregulation of LEAP2 during the postprandial state in mice. Finally, insulin and glucagon receptor-expressing hepatocytes are the primary source of hepatic LEAP2 expression, coinciding with a putative enhancer-like signature bound by insulin- and glucagon-regulated transcription factors at the LEAP2 locus. Collectively, our findings implicate insulin and glucagon in regulating LEAP2 and warrant further investigations into the exact mechanisms orchestrating this endocrine axis.
Collapse
Affiliation(s)
- Valdemar Brimnes Ingemann Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Diabetes Pharmacology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark; Diabetes and Metabolism Biology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Anna Katrina Jógvansdóttir Gradel
- Diabetes Pharmacology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark; Section of Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephanie Kjærulff Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joyceline Cuenco
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Merrild
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Natalia Petersen
- Diabetes and Metabolism Biology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Damien Demozay
- Diabetes and Metabolism Biology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Bharath Kumar Mani
- Obesity and NASH Research, Global Drug Discovery, Novo Nordisk, Lexington, MA, USA
| | - Malte Palm Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Magnus F G Grøndahl
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Asger Bach Lund
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Filip Krag Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Cesar A Prada-Medina
- Systems Biology and Target Discovery, AI and Digital Research, Novo Nordisk Research Center Oxford, Novo Nordisk A/S, Oxford, UK
| | | | - Jens Lykkesfeldt
- Section of Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Myrte Merkestein
- Diabetes Pharmacology, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Tatsuta R, Tanaka R, Tashibu A, Suzuki Y, Suzuki K, Shibata T, Ando T, Shin T, Sato Y, Itoh H. Association of chemotherapy-induced nausea and vomiting or anorexia with plasma levels of five gastrointestinal peptides in patients receiving chemotherapy. J Pharm Health Care Sci 2025; 11:17. [PMID: 40045433 PMCID: PMC11881272 DOI: 10.1186/s40780-025-00424-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Imbalance between gastrointestinal peptides has been implicated as a cause of chemotherapy-induced nausea and vomiting (CINV) and anorexia in cancer patients. This study comprehensively evaluated the changes in blood levels of five gastrointestinal peptide: substance P, neuropeptide (NPY), motilin, ghrelin and leptin, following chemotherapy, and the relationship between these peptides and CINV or anorexia. METHODS This single-center, prospective, observational study recruited 20 patients with esophageal cancer, urothelial cancer, or testiculoma undergoing cisplatin-based chemotherapy. Plasma levels of five gastrointestinal peptides were measured on days 1 (baseline; before administering chemotherapy), 3, 5 and 8 of the chemotherapy session. Anorexia and CINV were defined as visual analog scale scores 25 mm or higher at least once during the observation period. RESULTS Plasma NPY and leptin were significantly elevated in the early phase (day 3) of the chemotherapy session, while plasma motilin and substance P were significantly elevated in the late phase (days 5 and 8). Plasma motilin showed significant elevation on days 5 and 8 compared to baseline in CINV group but no significant increase in non-CINV group, and the levels were significantly higher in CINV than in non-CINV group. Plasma leptin peaked significantly on day 3 in both anorexia and non-anorexia groups, and remained significantly higher on day 5 compared to baseline in anorexia group but not in non-anorexia group. CONCLUSION CINV is associated with excessive secretion of motilin and anorexia is related to sustained elevation of leptin, suggesting the potential of these peptides as quantitative indicators of CINV and anorexia.
Collapse
Affiliation(s)
- Ryosuke Tatsuta
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan.
| | - Ryota Tanaka
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Asami Tashibu
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Yosuke Suzuki
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Kosuke Suzuki
- Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Tomotaka Shibata
- Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Tadasuke Ando
- Department of Urology, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Toshitaka Shin
- Department of Urology, Oita University Faculty of Medicine, Yufu, Oita, Japan
| | - Yuhki Sato
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| | - Hiroki Itoh
- Department of Clinical Pharmacy, Oita University Hospital, Yufu, Oita, Japan
| |
Collapse
|
3
|
Shiimura Y, Im D, Tany R, Asada H, Kise R, Kurumiya E, Wakasugi-Masuho H, Yasuda S, Matsui K, Kishikawa JI, Kato T, Murata T, Kojima M, Iwata S, Masuho I. The structure and function of the ghrelin receptor coding for drug actions. Nat Struct Mol Biol 2025; 32:531-542. [PMID: 39833471 DOI: 10.1038/s41594-024-01481-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Drugs targeting the ghrelin receptor hold therapeutic potential in anorexia, obesity and diabetes. However, developing effective drugs is challenging. To tackle this common issue across a broad drug target, this study aims to understand how anamorelin, the only approved drug targeting the ghrelin receptor, operates compared to other synthetic drugs. Our research elucidated the receptor's structure with anamorelin and miniGq, unveiling anamorelin's superagonistic activity. We demonstrated that ligands with distinct chemical structures uniquely bind to the receptor, resulting in diverse conformations and biasing signal transduction. Moreover, our study showcased the utility of structural information in effectively identifying natural genetic variations altering drug action and causing severe functional deficiencies, offering a basis for selecting the right medication on the basis of the individual's genomic sequence. Thus, by building on structural analysis, this study enhances the foundational framework for selecting therapeutic agents targeting the ghrelin receptor, by effectively leveraging signaling bias and genetic variations.
Collapse
Affiliation(s)
- Yuki Shiimura
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan.
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Dohyun Im
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Tany
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Hidetsugu Asada
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryoji Kise
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Eon Kurumiya
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | | | - Satoshi Yasuda
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
- Membrane Protein Research Center, Chiba University, Chiba, Japan
| | - Kazuma Matsui
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Jun-Ichi Kishikawa
- Institute for Protein Research, Osaka University, Osaka, Japan
- Faculty of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | - Takayuki Kato
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
- Membrane Protein Research Center, Chiba University, Chiba, Japan
| | - Masayasu Kojima
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Ikuo Masuho
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA.
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA.
| |
Collapse
|
4
|
Prost S, Elbers JP, Slezacek J, Hykollari A, Fuselli S, Smith S, Fusani L. The unexpected loss of the 'hunger hormone' ghrelin in true passerines: a game changer in migration physiology. ROYAL SOCIETY OPEN SCIENCE 2025; 12:242107. [PMID: 40109942 PMCID: PMC11919490 DOI: 10.1098/rsos.242107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Migratory birds must accumulate large amounts of fat prior to migration to sustain long flights. In passerines, the small body size limits the amount of energy stores that can be transported, and therefore birds undergo cycles of extreme fattening and rapid exhaustion of reserves. Research on these physiological adaptations was rattled by the discovery that birds have lost the main vertebrate regulator of fat deposition, leptin. Recent studies have thus focused on ghrelin, known as 'hunger hormone', a peptide secreted by the gastrointestinal tract to regulate, e.g. food intake and body mass in vertebrates. Studies on domestic species showed that, in birds, ghrelin has effects opposite to those described in mammals such as inhibiting instead of promoting food intake. Furthermore, recent studies have shown that ghrelin administration influences migratory behaviour in passerine birds. Using comparative genomics and immunoaffinity chromatography, we show that ghrelin has been lost in Eupasseres after the basic split from Acanthisitti about 50 Ma. We found that the ghrelin receptor is still conserved in passerines. The maintenance of a functional receptor system suggests that in Eupasserines, another ligand has replaced ghrelin, perhaps to bypass the feedback system that would hinder the large pre-migratory accumulation of subcutaneous fat.
Collapse
Affiliation(s)
- Stefan Prost
- Ecology and Genetics Research Unit, University of Oulu, Oulu, Finland
| | - Jean P Elbers
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Julia Slezacek
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Alba Hykollari
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Silvia Fuselli
- Life Sciences and Biotechnologies, University of Ferrara, Ferrara, Italy
| | - Steve Smith
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Leonida Fusani
- Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
- Department of Behavioral and Cognitive Biology, University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Edwards A, DeSante S, Spencer CD, Hyland L, Smith A, Sankhe AS, Szilvásy-Szabó A, Fekete C, Hill MN, Chee MJ, Abizaid A. Ghrelin Recruits the Endocannabinoid System to Modulate Food Reward. J Neurosci 2025; 45:e1620242024. [PMID: 39779372 DOI: 10.1523/jneurosci.1620-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/05/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Ghrelin enhances feeding by activating the growth hormone secretagogue receptor (GHSR). In the brain, GHSRs are expressed in regions responsible for regulating food motivation including the ventral tegmental area (VTA). Endogenous cannabinoids also promote food-seeking behaviors through the cannabinoid receptor-1 (CB-1Rs) in brain regions including the VTA. It is not known, however, if ghrelin and endocannabinoids interact in the VTA to produce these effects. We therefore examined if GHSR and CB-1R interact within the VTA to enhance food motivation. Results show that GHSR and CB-1R mRNA are expressed in the VTA cells in male and female rats and mice, with the GHSR being expressed in dopamine cells and the CB-1R being expressed primarily in nondopaminergic cells with no obvious sex differences. Ghrelin directly activated and increased excitatory tone onto dopamine cells of male and female mice. Male rats lacking fully functional GHSR signaling showed disrupted gene expression of transcripts important for regulating the synthesis, release, and degradation of endocannabinoids and lowered the levels of 2-arachidonoylglycerol (2-AG) within the VTA. Moreover, pharmacological antagonism of VTA CB-1Rs attenuates the orexigenic and appetitive effects of intra-VTA ghrelin in rats and blocks the ability of ghrelin to promote excitatory drive to VTA dopamine neurons. Finally, blocking the breakdown of cannabinoids in the VTA enhances the effects of ghrelin on food motivation. Together, our data show that ghrelin stimulates VTA dopamine cells and ultimately food motivation in part through a mechanism that involves endocannabinoid signaling at the CB-1R.
Collapse
Affiliation(s)
- Alexander Edwards
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Stephanie DeSante
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Carl Duncan Spencer
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Lindsay Hyland
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Andrea Smith
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Aditi S Sankhe
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Anett Szilvásy-Szabó
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest H-1083, Hungary
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest H-1083, Hungary
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N4T1, Canada
| | - Melissa J Chee
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Alfonso Abizaid
- Neuroscience Department, Carleton University, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
6
|
Okuro K, Hashimoto H, Morita K, Noshi Y, Kusushita M, Ochiai S, Utsu Y, Kasayama S, Hashimoto K. Anamorelin Induced Hyperglycemia in a Patient with Type 1 Diabetes by Stimulating Growth Hormone Secretion: A Case Report. Intern Med 2025:4887-24. [PMID: 39993750 DOI: 10.2169/internalmedicine.4887-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/26/2025] Open
Abstract
Anamorelin is a selective ghrelin receptor agonist that is used to treat cancer-related cachexia. Ghrelin stimulates growth hormone (GH) secretion. However, the association between anamorelin and hyperglycemia remains unclear. We herein report a case of anamorelin-induced hyperglycemia in a patient with type 1 diabetes mellitus. A 67-year-old woman with a history of type 1 diabetes was hospitalized because of a pleural effusion. After hospitalization, the patient was administered anamorelin. Four days after starting anamorelin treatment, her blood glucose and GH levels increased. These results suggest that anamorelin stimulates GH secretion and induces hyperglycemia. Our findings suggest the need to take special care of hyperglycemia when anamorelin is prescribed to patients with type 1 diabetes.
Collapse
|
7
|
Micarelli A, Mrakic-Sposta S, Vezzoli A, Malacrida S, Caputo S, Micarelli B, Misici I, Carbini V, Iennaco I, Granito I, Longo VD, Alessandrini M. Chemosensory and cardiometabolic improvements after a fasting-mimicking diet: A randomized cross-over clinical trial. Cell Rep Med 2025; 6:101971. [PMID: 39970875 DOI: 10.1016/j.xcrm.2025.101971] [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: 05/08/2024] [Revised: 09/26/2024] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Obesity is associated with a decrease in chemosensory perception acuity and increased disease risk, pointing to the need for feasible interventions that affect smell, taste, and cardiometabolic markers. Here, subjects with overweight/obesity are treated with six monthly cycles of a fasting-mimicking diet (FMD) lasting 5 days followed by a normal diet for the rest of the month to determine their effects on chemosensory function and cardiometabolic risk factors. Both arms of the 102 randomized cross-over trial participants indicate FMD-dependent improvements in a wide range of taste and smell chemosensory functions. The portion of hyposmic subjects is reduced from 38.1% at baseline to 6.4% at the end of 6 FMD cycles. FMD cycles also reduce cardiometabolic and inflammatory markers and drug use in diabetic patients. This trial provides evidence for the effect of periodic FMD cycles in improving chemosensory function while reducing cardiometabolic risk factors without requiring long-term lifestyle changes. The trial is registered at ClinicalTrials.gov (NCT04529161).
Collapse
Affiliation(s)
- Alessandro Micarelli
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy.
| | | | - Alessandra Vezzoli
- Institute of Clinical Physiology, National Research Council (CNR), Milan, Italy
| | - Sandro Malacrida
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
| | | | - Beatrice Micarelli
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Ilaria Misici
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Valentina Carbini
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Ilaria Iennaco
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Ivan Granito
- Unit of Neuroscience, Rehabilitation and Sensory Organs, UNITER ONLUS, Rome, Italy
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| | - Marco Alessandrini
- University of Rome Tor Vergata - Department of Clinical Sciences and Translational Medicine - Ear-Nose-Throat Unit, Rome, Italy
| |
Collapse
|
8
|
Ullah S, Feng F, Zhao M, Zhang J, Shao Q. Effect of dietary supplementation of lauric acid on growth performance, digestive enzymes, serum immune and antioxidant parameters, and intestinal morphology in black sea bream. FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:43. [PMID: 39918627 DOI: 10.1007/s10695-025-01457-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025]
Abstract
An eight-week feeding trial was conducted to examine the impact of dietary supplementation with lauric acid (LA) on juvenile black sea bream. A basal diet was formulated containing 19.9% fish meal, while five additional diets were prepared, each supplemented with varying levels of LA: LA1 (0.01%), LA2 (0.02%), LA3 (0.04%), LA4 (0.08%), and LA5 (0.16%), denoted as LA1 through LA5, respectively. Triplicate tanks were randomly allocated to each diet, each containing 20 fish with an initial weight of 1.55 ± 0.02 g. At the conclusion of the trial, the LA3 group exhibited significantly greater final body weight (FBW), weight gain (WG), specific growth rate (SGR), and protein efficiency ratio (PER) compared to the other groups (P < 0.05), while the feed conversion ratio (FCR) was markedly higher in the control group. No significant differences were detected among the groups in terms of initial body weight (IBW), muscle fiber index (MFI), protein productive value (PPV), condition factor (CF), hepatosomatic index (HSI), intraperitoneal fat (IPF), viscerosomatic index (VSI), and survival rate (SR) (P > 0.05). No significant variations were observed among the groups in the proximate compositions of the dorsal muscle and whole body (p > 0.05). Furthermore, no significant differences were observed in serum immune and antioxidant parameters in the midgut and hindgut and digestive enzyme activity (P > 0.05) among the treatment groups. However, the LA3 group demonstrated significantly higher levels of serum immune response markers IgM, C3, and C4 compared to the other groups, while malondialdehyde (MDA) levels were significantly elevated in the control group relative to the others. The LA3 group demonstrated significantly increased fore-intestinal villus height, crypt depth, villus height-to-crypt depth ratio, and goblet cell count per villus compared to the other groups (P < 0.05).
Collapse
Affiliation(s)
- Sami Ullah
- Zhejiang University Zhongyuan Institute, Zhengzhou, 450001, China
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Fengqin Feng
- Zhejiang University Zhongyuan Institute, Zhengzhou, 450001, China
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Minjie Zhao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jinzhi Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Qingjun Shao
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- Ocean Academy, Zhejiang University, Zhoushan, 316021, China.
| |
Collapse
|
9
|
Hankir MK, Lutz TA. Novel neural pathways targeted by GLP-1R agonists and bariatric surgery. Pflugers Arch 2025; 477:171-185. [PMID: 39644359 PMCID: PMC11761532 DOI: 10.1007/s00424-024-03047-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
The glucagon-like peptide 1 receptor (GLP-1R) agonist semaglutide has revolutionized the treatment of obesity, with other gut hormone-based drugs lined up that show even greater weight-lowering ability in obese patients. Nevertheless, bariatric surgery remains the mainstay treatment for severe obesity and achieves unparalleled weight loss that generally stands the test of time. While their underlying mechanisms of action remain incompletely understood, it is clear that the common denominator between GLP-1R agonists and bariatric surgery is that they suppress food intake by targeting the brain. In this Review, we highlight recent preclinical studies using contemporary neuroscientific techniques that provide novel concepts in the neural control of food intake and body weight with reference to endogenous GLP-1, GLP-1R agonists, and bariatric surgery. We start in the periphery with vagal, intestinofugal, and spinal sensory nerves and then progress through the brainstem up to the hypothalamus and finish at non-canonical brain feeding centers such as the zona incerta and lateral septum. Further defining the commonalities and differences between GLP-1R agonists and bariatric surgery in terms of how they target the brain may not only help bridge the gap between pharmacological and surgical interventions for weight loss but also provide a neural basis for their combined use when each individually fails.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
Alogaiel DM, Alsuwaylihi A, Alotaibi MS, Macdonald IA, Lobo DN. Effects of Ramadan intermittent fasting on hormones regulating appetite in healthy individuals: A systematic review and meta-analysis. Clin Nutr 2025; 45:250-261. [PMID: 39842253 DOI: 10.1016/j.clnu.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/17/2024] [Accepted: 01/05/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND AND AIMS This systematic review and meta-analysis aimed to examine the effect of Ramadan intermittent fasting on appetite-regulating hormones including leptin, ghrelin, insulin, gastrin, glucagon-like peptide-1, peptide YY, and cholecystokinin. METHODS We searched the MEDLINE, Embase, Cochrane Library, CINAHL, Google Scholar, and Web of Science databases to identify relevant research on appetite-regulating hormones during Ramadan intermittent fasting, published until the end of March 2024. RESULTS Data from 16 eligible studies comprising 664 participants (341, 51.4 % male) with a mean ± standard deviation age of 33.9 ± 10.8 years were included. The meta-analysis included 12 studies with complete leptin data, showing no significant effect of Ramadan intermittent fasting on leptin concentrations (standardised mean difference - SMD = -0.11 μg/mL, 95 % CI: -0.36 to 0.14). Analysis of three studies with complete ghrelin data demonstrated a significant increase in ghrelin concentrations following Ramadan intermittent fasting (SMD = 0.31 pg/mL, 95 % CI: 0.03 to 0.60). Six studies examining insulin concentrations pre- and post-fasting revealed no significant effect on insulin concentrations (SMD = -0.24 μU/mL, 95 % CI: -0.54 to 0.02). Similarly, analysis of three studies with complete gastrin data showed no significant effect of intermittent fasting on gastrin concentrations (SMD = 0.23 pg/mL, 95 % CI: -0.71 to 0.99). CONCLUSION Ramadan intermittent fasting significantly increases ghrelin concentrations while showing no significant effects on leptin, insulin, and gastrin. While ghrelin findings were consistent across studies, the high heterogeneity in leptin studies suggests further research to better understand the effects of Ramadan intermittent fasting on appetite-regulating hormones.
Collapse
Affiliation(s)
- Deema M Alogaiel
- Nottingham Digestive Diseases Centre, Division of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK; Health Sciences Department, College of Health and Rehabilitation, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Abdulaziz Alsuwaylihi
- Nottingham Digestive Diseases Centre, Division of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; Department of Clinical Nutrition, King Saud Medical City, Ministry of Health, Riyadh, Saudi Arabia
| | - May S Alotaibi
- Health Sciences Department, College of Health and Rehabilitation, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia; Division of Food, Nutrition & Dietetics, School of Biosciences, University of Nottingham, LE12 5RD, UK
| | - Ian A Macdonald
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Dileep N Lobo
- Nottingham Digestive Diseases Centre, Division of Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham, UK; MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK; Division of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Cuberos Paredes E, Goyes D, Mak S, Yardimian R, Ortiz N, McLaren A, Stauss HM. Transcutaneous auricular vagus nerve stimulation inhibits mental stress-induced cortisol release-Potential implications for inflammatory conditions. Physiol Rep 2025; 13:e70251. [PMID: 39936474 DOI: 10.14814/phy2.70251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Elevated glucocorticoid levels with reduced glucocorticoid responsiveness have been reported in chronic inflammatory conditions. Activation of neurons in the nucleus of the solitary tract by transcutaneous auricular vagus nerve stimulation (taVNS) may activate inhibitory pathways projecting to the hypothalamic paraventricular nucleus (PVN), thus inhibiting corticotropin-releasing hormone (CRH) release and improving glucocorticoid dysfunction in chronic inflammatory conditions. Healthy adults (n = 12) participated in experimental (taVNS) and control (sham-taVNS) sessions at least 4 days apart. A 30-min baseline recording was followed by 30 min of taVNS or sham-taVNS and 40 min of recovery. Ten minutes into taVNS or sham-taVNS, a mental arithmetic stress test (MAST) was conducted for 15 min. The MAST increased heart rate, low frequency (LF) heart rate variability (HRV), and the LF to high frequency ratio of HRV, confirming sympathetic activation. Salivary cortisol levels during the MAST were lower during taVNS (49.5 ± 48.0% from baseline; mean ± SD) compared to sham-taVNS (106.0 ± 81.1% from baseline; mean ± SD; p < 0.05). In a psoriasis patient, daily taVNS for 3 months reduced diurnal salivary cortisol levels from 58.2 ± 35.2 (ng/mL)*h (mean ± SD) to 34.9 ± 13.8 (ng/mL)*h (mean ± SD). While it is possible that taVNS inhibited CRH-releasing neurons in the PVN, our study design did not allow to confirm this potential mechanism.
Collapse
Affiliation(s)
- Ely Cuberos Paredes
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Domenica Goyes
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Sadie Mak
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Raffi Yardimian
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Nickolas Ortiz
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Ayana McLaren
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Harald M Stauss
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| |
Collapse
|
12
|
Sixten HJ, Rønnestad I, Bogevik AS, Aspevik T, Oterhals Å, Gomes AS, Lai F, Tolås I, Gelebard V, Hillestad M, Kousoulaki K. Side-Stream Based Marine Solubles From Atlantic Cod ( Gadus morhua) Modulate Appetite and Dietary Nutrient Utilization in Atlantic Salmon ( Salmo salar L.) and can Replace Fish Meal. AQUACULTURE NUTRITION 2025; 2025:4872889. [PMID: 39949357 PMCID: PMC11824393 DOI: 10.1155/anu/4872889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025]
Abstract
Whitefish fisheries' side-stream biomass is an abundant underutilized resource that can be valorized to benefit future aquaculture sustainability. Four novel ingredients based on side-streams from Atlantic cod (Gadus morhua) fileting were produced. FM-hb, a fish meal (FM), and FPH-hb, a fish protein hydrolysate based on heads (h) and backbones (b); FM-hbg, a FM based on heads, backbones, and viscera/guts (g); and FPC-g, a fish protein concentrate based on viscera preserved in formic acid. Four diets were prepared containing one of the ingredients replacing 50% of the dietary FM protein, in addition to a positive (FM10) and a negative (FM5) control. The six diets were fed to triplicate tanks with Atlantic salmon (Salmo salar L.; 113 ± 1 g) over 8 weeks. Besides general performance, gut and brain gene expression for selected hormones and key neuropeptides involved in the control of appetite and digestive processes were studied during feeding and postprandial, and possible reference levels for Atlantic salmon were established. All side-stream-added diets performed well, with no significant differences in performance and biometrics between the treatments. Some gene expression differences were observed, but no well-defined patterns emerged supporting clear dietary effects related to digestive performance or appetite. However, in the brain, a short-time upregulation of agouti-related protein-1 (agrp1), corresponded to higher cumulative feed intake (FI) for the FM10 diet supporting notions that this may be a candidate biomarker for appetite in salmon. Expression of stomach ghrelin-1 (ghrl1) was higher than ghrelin-2 (ghrl2) and membrane-bound O-acyltransferase domain-containing 4 (mboat4), and midgut peptide YYa-2 (pyya2) and glucagon-a (gcga) were higher than peptide YYb-1 (pyyb1). A comparison showed that midgut peptide YYa-1 (pyya1), pyya2, and gcga expressions were higher than in the hindgut, which is opposite of what is found in mammals. In conclusion, this study shows that sustainable side-stream raw materials with different characteristics can partly replace high-quality commercial FMs giving similar performance.
Collapse
Affiliation(s)
- Hanne Jorun Sixten
- Department of Research and Development, BioMar AS, Trondheim, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Ivar Rønnestad
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - André S. Bogevik
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Tone Aspevik
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Åge Oterhals
- Department of Nutrition and Feed Technology, NOFIMA, Fyllingsdalen, Norway
| | - Ana S. Gomes
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Institute of Marine Research, Tromsø, Norway
| | - Floriana Lai
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Ingvill Tolås
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Department of Biological Sciences, NTNU Ålesund, Ålesund, Norway
| | - Virginie Gelebard
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Marie Hillestad
- Department of Research and Development, BioMar AS, Trondheim, Norway
| | | |
Collapse
|
13
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
14
|
Warwas N, Langeland M, Roques JAC, Montjouridès M, Smeets J, Sundh H, Jönsson E, Sundell K. Fish processing side streams are promising ingredients in diets for rainbow trout (Oncorhynchus mykiss) -Effects on growth physiology, appetite, and intestinal health. JOURNAL OF FISH BIOLOGY 2025; 106:75-92. [PMID: 37843903 PMCID: PMC11758193 DOI: 10.1111/jfb.15589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
Due to the growth of aquaculture and the finite supply of fishmeal and oil, alternative marine protein and lipid sources are highly sought after. Particularly promising is the use of side streams from the fish processing industry, allowing for the recovery and retention of otherwise lost nutrients in the food production chain. The aim of the present study was to evaluate the potential of three fish processing side streams as fish feed ingredients. The side streams originated from different stages of the production chain, were used without further processing, and included sprat trimmings (heads, frames, viscera), marinated herring (fillets), and mackerel in tomato sauce (fillets and sauce). The three side streams contained moderate levels of protein (28-32% dry matter) and high levels of lipid (34-43%). The sprat trimmings included ca. 29% ash and 1.5% phosphorous, which may add value due to the high level of essential minerals but needs to be considered in feed formulations. Three diets were formulated to include 50% of each side stream replacing all fishmeal and ca. 80% of the fish oil of the control diet, which contained 35% fishmeal and 10% fish oil. The diets were evaluated in a 12-week feeding trial using rainbow trout (Oncorhynchus mykiss). Fish fed the sprat diet displayed the highest feed intake and growth and showed no negative effects on the intestinal health. The mackerel side stream displayed a good digestibility but resulted in lower growth rates compared to the sprat trimmings. Fish fed the herring diet displayed the lowest performance regarding growth, feed intake, and digestibility. They further exhibited a reduction in nutrient uptake in both proximal and distal intestines, likely contributing to the observed lower digestibility and growth, and a reduction in plasma ghrelin levels. As part of a circular approach to increase marine lipid and protein production for fish feed, the tested sprat and mackerel side streams are promising raw materials; however, additional studies using more commercial-like feed formulations are encouraged.
Collapse
Affiliation(s)
- Niklas Warwas
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
- Swedish Mariculture Research CenterSWEMARC, University of GothenburgGothenburgSweden
- Blue Food, Center for Future SeafoodUniversity of GothenburgGothenburgSweden
| | - Markus Langeland
- Blue Food, Center for Future SeafoodUniversity of GothenburgGothenburgSweden
- RISE Research Institute of SwedenGothenburgSweden
| | - Jonathan A. C. Roques
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
- Swedish Mariculture Research CenterSWEMARC, University of GothenburgGothenburgSweden
| | - Marie Montjouridès
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
| | - Jolie Smeets
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
| | - Henrik Sundh
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
- Swedish Mariculture Research CenterSWEMARC, University of GothenburgGothenburgSweden
| | - Elisabeth Jönsson
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
- Swedish Mariculture Research CenterSWEMARC, University of GothenburgGothenburgSweden
- Blue Food, Center for Future SeafoodUniversity of GothenburgGothenburgSweden
| | - Kristina Sundell
- Department of Biological and Environmental SciencesUniversity of GothenburgGothenburgSweden
- Swedish Mariculture Research CenterSWEMARC, University of GothenburgGothenburgSweden
- Blue Food, Center for Future SeafoodUniversity of GothenburgGothenburgSweden
| |
Collapse
|
15
|
Urai H, Azegami T, Komatsu M, Takahashi R, Kubota Y, Hasegawa K, Tokuyama H, Wakino S, Hayashi K, Kanda T, Itoh H. Ghrelin Promotes Lipid Uptake into White Adipose Tissue via Endothelial Growth Hormone Secretagogue-Receptor in Mice. Nutrients 2024; 17:146. [PMID: 39796581 PMCID: PMC11722803 DOI: 10.3390/nu17010146] [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: 12/14/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Endothelial peroxisome proliferator-activated receptor gamma (PPARγ) regulates adipose tissue by facilitating lipid uptake into white adipocytes, but the role of endothelial lipid transport in systemic energy balance remains unclear. Ghrelin conveys nutritional information through the central nervous system and increases adiposity, while deficiency in its receptor, growth hormone secretagogue-receptor (GHSR), suppresses adiposity on a high-fat diet. This study aims to examine the effect of ghrelin/GHSR signaling in the endothelium on lipid metabolism. Methods: We compared the effects of ghrelin on adiposity and lipid uptake into adipocytes in wild-type and GHSR-null mice. Transgenic mice expressing GHSR selectively in endothelial cells were also generated and compared with global GHSR-null and wild-type mice. The impact of ghrelin on lipid uptake-related genes was assessed in cultured endothelial cells. Results: Ghrelin increased adiposity and triglyceride clearance in wild-type but not in GHSR-null mice. GHSR-null mice showed higher serum triglyceride after olive oil gavage and lower white adipose tissue (WAT) weight on a high-fat diet, suggesting impaired lipid uptake. Restoring GHSR expression in endothelial cells increased lipoprotein lipase activity, lipid uptake into WAT, and WAT weight. Ghrelin enhanced free fatty acid uptake and the expression of lipid uptake genes in cultured endothelial cells, whereas these effects were absent in GHSR-null mice-derived endothelial cells. Knockdown of PPARγ revealed that ghrelin/GHSR signaling in endothelial cells promoted lipid uptake via endothelial PPARγ. Conclusions: Endothelial GHSR is key for regulating lipid metabolism via PPARγ in response to ghrelin and for the role of endothelium in regulating white adipocyte metabolism. Targeting endothelial ghrelin signaling may be a promising therapeutic approach for managing excessive adiposity and associated metabolic disorders.
Collapse
Affiliation(s)
- Hidenori Urai
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; (H.U.); (M.K.); (R.T.); (K.H.); (H.I.)
| | - Tatsuhiko Azegami
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; (H.U.); (M.K.); (R.T.); (K.H.); (H.I.)
| | - Motoaki Komatsu
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; (H.U.); (M.K.); (R.T.); (K.H.); (H.I.)
| | - Rina Takahashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; (H.U.); (M.K.); (R.T.); (K.H.); (H.I.)
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Kazuhiro Hasegawa
- Division of Nephrology, Department of Internal Medicine, Tokushima University School of Medicine, Tokushima-shi 770-8503, Tokushima, Japan; (K.H.); (S.W.)
| | - Hirofumi Tokuyama
- Department of Internal Medicine, Tokyo Dental University Ichikawa General Hospital, Ichikawa-shi 272-8513, Chiba, Japan;
| | - Shu Wakino
- Division of Nephrology, Department of Internal Medicine, Tokushima University School of Medicine, Tokushima-shi 770-8503, Tokushima, Japan; (K.H.); (S.W.)
| | - Kaori Hayashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; (H.U.); (M.K.); (R.T.); (K.H.); (H.I.)
| | - Takeshi Kanda
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; (H.U.); (M.K.); (R.T.); (K.H.); (H.I.)
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Matsue 693-8501, Shimane, Japan
- The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Matsue 693-8501, Shimane, Japan
| | - Hiroshi Itoh
- Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan; (H.U.); (M.K.); (R.T.); (K.H.); (H.I.)
| |
Collapse
|
16
|
Faulkner ML, Farokhnia M, Lee MR, Farinelli L, Browning BD, Abshire K, Daurio AM, Munjal V, Deschaine SL, Boukabara SR, Fortney C, Sherman G, Schwandt M, Akhlaghi F, Momenan R, Ross TJ, Persky S, Leggio L. A randomized, double-blind, placebo-controlled study of a GHSR blocker in people with alcohol use disorder. JCI Insight 2024; 9:e182331. [PMID: 39704175 DOI: 10.1172/jci.insight.182331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUNDStudies have demonstrated the role of ghrelin in alcohol-related behaviors and consumption. Blockade of the growth hormone secretagogue receptor (GHSR), which is the ghrelin receptor, has been shown to decrease alcohol drinking and reward-related behaviors across several animal models. We previously conducted a human study testing a GHSR inverse agonist/competitive antagonist, PF-5190457, in individuals who are heavy drinkers and showed its safety when coadministered with alcohol. Here, we conducted a phase IIa experimental medicine study in patients with alcohol use disorder (AUD) to investigate the effects of PF-5190457 on alcohol- and food-related outcomes.METHODSForty-two individuals with AUD (n = 29 completers) participated in a randomized, double-blind, placebo-controlled study where they received PF-5190457 100mg b.i.d. (or placebo) in 2 counterbalanced, within-subject stages. Participants completed an alcohol cue-reactivity (CR) experiment in a bar-like laboratory and a virtual food choice experiment in a cafeteria-like virtual reality (VR) environment. A subset of participants (n = 12) performed a CR task during a brain functional MRI (fMRI) experiment.RESULTSPF-5190457 did not reduce cue-elicited alcohol craving. PF-5190457 reduced virtual calories selected (P = 0.04) in the VR environment. PF-5190457 did not influence neural activation during CR task in the fMRI experiment.CONCLUSIONThis study provides human evidence of the role of GHSR blockade in behaviors related to food selection and highlights the need for future investigations into targeting the ghrelin system in AUD.TRIAL REGISTRATIONClinicalTrials.gov (accession no. NCT02707055).FUNDINGNIDA and NIAAA ZIA-DA000635; National Center for Advancing Translational Sciences UH2/UH3-TR000963.
Collapse
Affiliation(s)
- Monica L Faulkner
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mary R Lee
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Lisa Farinelli
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Brittney D Browning
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Kelly Abshire
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Allison M Daurio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Vikas Munjal
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Sara L Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Selim R Boukabara
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Christopher Fortney
- Immersive Simulation Program, Social and Behavioral Research Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Garrick Sherman
- Office of the Clinical Director, NIDA, Intramural Research Program, NIH, Baltimore, Maryland, USA
| | - Melanie Schwandt
- Office of the Clinical Director, NIAAA Division of Intramural Clinical and Biological Research, NIH, Bethesda, Maryland, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Reza Momenan
- Clinical NeuroImaging Research Core, NIAAA, NIH, Bethesda, Maryland, USA
| | - Thomas J Ross
- Neuroimaging Core, NIDA Intramural Research Program, NIH, Baltimore, Maryland, USA
| | - Susan Persky
- Immersive Simulation Program, Social and Behavioral Research Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, 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
| |
Collapse
|
17
|
Recinella L, Libero ML, Brunetti L, Acquaviva A, Chiavaroli A, Orlando G, Granata R, Salvatori R, Leone S. Effects of growth hormone-releasing hormone deficiency in mice beyond growth. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09936-3. [PMID: 39695049 DOI: 10.1007/s11154-024-09936-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
This paper provides a critical overview on GHRH and its deficiency, discussing its multiple roles in both central and peripheral tissues. Genetically engineered mice have been instrumental in elucidating the multifaceted roles of GHRH and GH, each offering unique insights into the physiological and pathological roles of these hormones, although in many of these models dissecting the direct effect of GHRH from the effect of GH is not possible. Key findings highlight the effects of GHRH deficiency on emotional behavior, including anxiety and depression, its impact on memory and learning capabilities, as well as on adipose tissue, immune system, inflammation and pain.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Maria Loreta Libero
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Luigi Brunetti
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy.
| | - Alessandra Acquaviva
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Annalisa Chiavaroli
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Giustino Orlando
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Riccarda Granata
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Roberto Salvatori
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sheila Leone
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
18
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [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: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
19
|
Bioletto F, Varaldo E, Gasco V, Maccario M, Arvat E, Ghigo E, Grottoli S. Central and peripheral regulation of the GH/IGF-1 axis: GHRH and beyond. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09933-6. [PMID: 39579280 DOI: 10.1007/s11154-024-09933-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
The regulation of growth hormone (GH) synthesis and secretion by somatotroph cells of the anterior pituitary is a highly complex process, mediated by a variety of neuroendocrine and peripheral influences. In particular, a key role is played by the hypothalamic peptides growth hormone-releasing hormone (GHRH) and somatostatin, which regulate the somatotroph axis with opposite actions, stimulating and inhibiting GH release, respectively. Since the discovery of GHRH about 50 years ago, many pathophysiological studies have explored the underlying intricate hormonal balance that regulates GHRH secretion and its interplay with the somatotroph axis. Various molecules and pathophysiological states have been shown to modulate the release of GH, GHRH, somatostatin and GH secretagogues. Collectively, the available evidence demonstrates how a vast number of neural and peripheral signals are conveyed and integrated to orchestrate a finely tuned response of the somatotroph axis that adapts to the body's varying needs for growth, metabolism, and repair. The present review aims to summarize the available evidence regarding the key regulators involved in the modulation of the somatotroph axis in humans, presenting detailed molecular insights on the signaling cascades at play. The interplay between different mechanisms governing somatotroph secretion is highlighted, underscoring the nuanced interdependence that maintains homeostasis and facilitates the body's ability to respond to internal and external stimuli.
Collapse
Affiliation(s)
- Fabio Bioletto
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Emanuele Varaldo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Valentina Gasco
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Mauro Maccario
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Emanuela Arvat
- Division of Oncological Endocrinology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Ezio Ghigo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Grottoli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
20
|
Fu ZT, Liu CZ, Kim MR, Liu YD, Wang Y, Fu YM, Yang JW, Yang NN. Acupuncture improves the symptoms, serum ghrelin, and autonomic nervous system of patients with postprandial distress syndrome: a randomized controlled trial. Chin Med 2024; 19:162. [PMID: 39568071 PMCID: PMC11580632 DOI: 10.1186/s13020-024-01028-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Whether gastrointestinal hormones in patients with postprandial distress syndrome (PDS) are altered by acupuncture, and whether such alterations are related to the autonomic nervous system (ANS), remains an open question. OBJECTIVE This study aims to investigate the effects of acupuncture on symptoms, serum hormones, and ANS in PDS patients. METHODS This randomized controlled clinical trial was conducted at Beijing Hospital of Traditional Chinese Medicine affiliated with Capital Medical University. Sixty-two PDS patients were randomly assigned equally to acupuncture or sham acupuncture arm (3 sessions per week for 4-week). The main outcome measures which were evaluated at baseline and 4-week included cardinal symptoms, serum hormones including ghrelin, vasoactive intestinal peptide (VIP), substance P (SP), and ANS. RESULTS Among the 62 randomly assigned participants, 51 (82%) were included in the baseline characteristics and outcome analysis. Gastrointestinal symptoms including response rate (p = 0.001) and dyspepsia symptom severity (p = 0.002) were significantly improved after acupuncture treatment. Serum ghrelin concentration was significantly higher in acupuncture group than in sham acupuncture group (8.34 ± 3.00 ng/ml versus 6.52 ± 2.00 ng/ml, p = 0.022) after 4-week treatment, instead of VIP and SP (p > 0.05). The acupuncture group had significantly higher vagal activity, showing with increasing of high-frequency component (HF, p ≤ 0.001) and decreasing of the ratio of low-frequency and HF (p ≤ 0.001). In relationship analysis, the HF component exhibited a significant inverse correlation with symptom severity (R = - 0.501, p ≤ 0.001), but not with ghrelin level (R = 0.026, p = 0.865). CONCLUSION Acupuncture may improve the symptoms and increase the ghrelin level of PDS patients, the therapeutic effect of acupuncture was associated with the alteration of vagal activity. TRIAL REGISTRATION The trial is registered with the ISRCTN registry, ISRCTN12511434. Registered 31 March 2017, https://www.isrctn.com/ .
Collapse
Affiliation(s)
- Zi-Tong Fu
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Cun-Zhi Liu
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Mi-Rim Kim
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Yi-Duo Liu
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Yu Wang
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Yi-Ming Fu
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Jing-Wen Yang
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Na-Na Yang
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, No. 11 Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
21
|
De Cristofaro M, Lenzi A, Ghimenti S, Biagini D, Bertazzo G, Vivaldi FM, Armenia S, Pugliese NR, Masi S, Di Francesco F, Lomonaco T. Decoding the Challenges: navigating Intact Peptide Mass Spectrometry-Based Analysis for Biological Applications. Crit Rev Anal Chem 2024:1-23. [PMID: 39556023 DOI: 10.1080/10408347.2024.2427140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Quantitative analysis of peptides in biological fluids offers a high diagnostic and prognostic tool to reflect the pathophysiological condition of the patient. Recently, methods based on liquid chromatography coupled with mass spectrometry (LC-MS) for the quantitative determination of intact peptides have been replacing traditionally used ligand-binding assays, which suffer from cross-reactivity issues. The use of "top-down" analysis of peptides is rapidly increasing since it does not undergo incomplete or non-reproducible digestion like "bottom-up" approaches. However, the low abundance of peptides and their peculiar characteristics, as well as the complexity of biological fluids, make their quantification challenging. Herein, the analytical pitfalls that may be encountered during the development of an LC-MS method for the analysis of intact peptides in biological fluids are discussed. Challenges in the pre-analytical phase, stability after sampling and sample processing, significantly impact the accuracy of peptide quantification. Emerging techniques, such as microextractions, are becoming crucial for improved sample cleanup and enrichment of target analytes. A comparison between the roles of high-resolution and low-resolution mass spectrometry in the quantification of intact peptides, as well as the introduction of supercharging reagents to enhance ionization, will be discussed.
Collapse
Affiliation(s)
| | - Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Silvia Ghimenti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Giulia Bertazzo
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | | | - Silvia Armenia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| |
Collapse
|
22
|
Huang L, Liu P, Du Y, Bazan JF, Pan D, Chen Q, Lee A, Kola VSR, Wolfe SA, Wang YX. A brown fat-enriched adipokine, ASRA, is a leptin receptor antagonist that stimulates appetite. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.12.557454. [PMID: 37745491 PMCID: PMC10515849 DOI: 10.1101/2023.09.12.557454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The endocrine control of food intake remains incompletely understood, and whether the leptin receptor (LepR)-mediated anorexigenic pathway in the hypothalamus is negatively regulated by a humoral factor is unknown. Here, we identify an appetite-stimulating factor - ASRA - that represents a peripheral signal of energy deficit and orthosterically antagonizes LepR signaling. Asra encodes an 8 kD protein that is abundantly and selectively expressed in adipose tissue and to a lesser extent, in liver. ASRA associates with autophagy vesicles and its secretion is enhanced by energy deficiency. In vivo, fasting and cold stimulate Asra expression and increase its protein concentration in cerebrospinal fluid. Asra overexpression attenuates LepR signaling, leading to elevated blood glucose and development of severe hyperphagic obesity. Conversely, either adipose- or liver-specific Asra knockout mice display increased leptin sensitivity, improved glucose homeostasis, reduced food intake, resistance to high-fat diet-induced obesity, and blunted cold-evoked feeding response. Mechanistically, ASRA acts as a high affinity antagonist of LepR. AlphaFold2-multimer prediction and mutational studies suggest that a core segment of ASRA binds to the immunoglobin-like domain of LepR, similar to the 'site 3' recognition of the A-B loop of leptin. While administration of recombinant wild-type ASRA protein promotes food intake and increases blood glucose in a LepR signaling-dependent manner, point mutation within ASRA that disrupts LepR-binding results in a loss of these effects. Our studies reveal a previously unknown endocrine mechanism in appetite regulation and have important implications for our understanding of leptin resistance.
Collapse
Affiliation(s)
- Lei Huang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - Pengpeng Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - Yong Du
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - J Fernando Bazan
- Bioconsulting llc, Stillwater, MN, USA
- Unit of Structural Biology, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Dongning Pan
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Present address: Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Qingbo Chen
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexandra Lee
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vijaya Sudhakara Rao Kola
- Department of Medicine and Division of Hematology/Oncology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yong-Xu Wang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
23
|
Zhang Y, Wang R, Liu T, Wang R. Exercise as a Therapeutic Strategy for Obesity: Central and Peripheral Mechanisms. Metabolites 2024; 14:589. [PMID: 39590824 PMCID: PMC11596326 DOI: 10.3390/metabo14110589] [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: 09/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a complex, multifactorial condition involving excessive fat accumulation due to an imbalance between energy intake and expenditure, with its global prevalence steadily rising. This condition significantly increases the risk of chronic diseases, including sarcopenia, type 2 diabetes, and cardiovascular diseases, highlighting the need for effective interventions. Exercise has emerged as a potent non-pharmacological approach to combat obesity, targeting both central and peripheral mechanisms that regulate metabolism, energy expenditure, and neurological functions. In the central nervous system, exercise influences appetite, mood, and cognitive functions by modulating the reward system and regulating appetite-controlling hormones to manage energy intake. Concurrently, exercise promotes thermogenesis in adipose tissue and regulates endocrine path-ways and key metabolic organs, such as skeletal muscle and the liver, to enhance fat oxidation and support energy balance. Despite advances in understanding exercise's role in obesity, the precise interaction between the neurobiological and peripheral metabolic pathways remains underexplored, particularly in public health strategies. A better understanding of these interactions could inform more comprehensive obesity management approaches by addressing both central nervous system influences on behavior and peripheral metabolic regulation. This review synthesizes recent insights into these roles, highlighting potential therapeutic strategies targeting both systems for more effective obesity interventions.
Collapse
Affiliation(s)
- Yiyin Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Ruwen Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| |
Collapse
|
24
|
Zhang J, Zhao Y, Wu S, Han M, Gao L, Yang K, Chen H, Wang C, Xu G. Mechanosensing by Piezo1 in gastric ghrelin cells contributes to hepatic lipid homeostasis in mice. Sci Signal 2024; 17:eadq9463. [PMID: 39436995 DOI: 10.1126/scisignal.adq9463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024]
Abstract
Ghrelin is an orexigenic peptide released by gastric ghrelin cells that contributes to obesity and hepatic steatosis. The mechanosensitive ion channel Piezo1 in gastric ghrelin cells inhibits the synthesis and secretion of ghrelin in response to gastric mechanical stretch. We sought to modulate hepatic lipid metabolism by manipulating Piezo1 in gastric ghrelin cells. Mice with a ghrelin cell-specific deficiency of Piezo1 (Ghrl-Piezo1-/-) had hyperghrelinemia and hepatic steatosis when fed a low-fat or high-fat diet. In these mice, hepatic lipid accumulation was associated with changes in gene expression and in protein abundance and activity expected to increase hepatic fatty acid synthesis and decrease lipid β-oxidation. Pharmacological inhibition of the ghrelin receptor improved hepatic steatosis in Ghrl-Piezo1-/- mice, thus confirming that the phenotype of these mice was due to overproduction of ghrelin caused by inactivation of Piezo1. Gastric implantation of silicone beads to induce mechanical stretch of the stomach inhibited ghrelin synthesis and secretion, thereby helping to suppress fatty liver development induced by a high-fat diet in wild-type mice but not in Ghrl-Piezo1-/- mice. Our study elucidates the mechanism by which Piezo1 in gastric ghrelin cells regulate hepatic lipid accumulation, providing insights into potential treatments for fatty liver.
Collapse
Affiliation(s)
- Jinshan Zhang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
- Department of Metabolic and Bariatric Surgery, First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Yawen Zhao
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Shaohong Wu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Mengxue Han
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Luyang Gao
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Ke Yang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Hui Chen
- Biotherapy Center; Cell-gene Therapy Translational Medicine Research Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, 510632 Guangzhou, Guangdong, China
| |
Collapse
|
25
|
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
|
26
|
Matsui K, Ida T, Oishi K, Kojima M, Sato T. Ghrelin is essential for lowering blood pressure during torpor. Front Endocrinol (Lausanne) 2024; 15:1487028. [PMID: 39449746 PMCID: PMC11499174 DOI: 10.3389/fendo.2024.1487028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Daily torpor is an active hypothermic phenomenon that is observed in some mammals and birds during fasting. A decrease in blood pressure has also been observed in torpor; however, there remains a lack of knowledge of the underlying mechanism. We have previously reported that ghrelin, an orexigenic hormone, has a hypothermic effect and is essential for the induction and maintenance of torpor. It is also known that the ghrelin secretion is enhanced during fasting and that ghrelin receptors are distributed in the cardiovascular system. Therefore, this study was conducted to test the hypothesis that ghrelin is actively involved in the regulation of blood pressure during torpor induction. Methods Male wild-type and ghrelin gene-deficient mice were generated by homologous recombination as previously reported. Mice, 10 weeks old, were included in this study and housed five per cage. The mice were maintained on a 12-h light/dark cycle (lights on from 7:00 to 19:00) with access to food and water ad libitum. Results The continuous measurement of blood pressure using a telemetry system showed that induction of torpor by fasting did not decrease blood pressure in ghrelin gene-deficient mice. The analysis of heart rate variability revealed that sympathetic nerve activity was predominant in ghrelin-deficient mice during fasting. Furthermore, these features were cancelled by administration of a ghrelin receptor agonist and were comparable to those in wild-type mice. Discussion In this study, we showed that blood pressure was elevated in ghrl-/- mice and that the blood pressure rhythm was abnormal. Furthermore, we showed that the ghrelin gene deficiency does not cause sufficient blood pressure reduction upon entry into the torpor, and that the administration of the ghrelin receptor agonist, GHRP-6, causes blood pressure reduction associated with torpor. Thus, we have shown for the first time that the active role of ghrelin is essential for active blood pressure reduction associated with torpor, and that this action is mediated by the inhibition of sympathetic nerve activity by ghrelin.
Collapse
Affiliation(s)
- Kazuma Matsui
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Takanori Ida
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Kanae Oishi
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Masayasu Kojima
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Takahiro Sato
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| |
Collapse
|
27
|
Yu J, Gao M, Wang L, Guo X, Liu X, Sheng M, Cheng S, Guo Y, Wang J, Zhao C, Guo W, Zhang Z, Liu Y, Hu C, Ma X, Xie C, Zhang Q, Xu L. An insoluble cellulose nanofiber with robust expansion capacity protects against obesity. Int J Biol Macromol 2024; 277:134401. [PMID: 39097049 DOI: 10.1016/j.ijbiomac.2024.134401] [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: 01/12/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
An imbalance between energy intake and energy expenditure predisposes obesity and its related metabolic diseases. Soluble dietary fiber has been shown to improve metabolic homeostasis mainly via microbiota reshaping. However, the application and metabolic effects of insoluble fiber are less understood. Herein, we employed nanotechnology to design citric acid-crosslinked carboxymethyl cellulose nanofibers (CL-CNF) with a robust capacity of expansion upon swelling. Supplementation with CL-CNF reduced food intake and delayed digestion rate in mice by occupying stomach. Besides, CL-CNF treatment mitigated diet-induced obesity and insulin resistance in mice with enhanced energy expenditure, as well as ameliorated inflammation in adipose tissue, intestine and liver and reduced hepatic steatosis, without any discernible signs of toxicity. Additionally, CL-CNF supplementation resulted in enrichment of probiotics such as Bifidobacterium and decreased in the relative abundances of deleterious microbiota expressing bile salt hydrolase, which led to increased levels of conjugated bile acids and inhibited intestinal FXR signaling to stimulate the release of GLP-1. Taken together, our findings demonstrate that CL-CNF administration protects mice from diet-induced obesity and metabolic dysfunction by reducing food intake, enhancing energy expenditure and remodeling gut microbiota, making it a potential therapeutic strategy against metabolic diseases.
Collapse
Affiliation(s)
- Jian Yu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyuan Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaodi Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Maozheng Sheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shimiao Cheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yingying Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiawen Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Cheng Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wenxiu Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhe Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yameng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xinran Ma
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China; Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, China.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Qiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
28
|
Omori T, Yamamoto K, Kurokawa Y, Miyazaki Y, Fujitani K, Kawabata R, Imamura H, Takeno A, Yanagimoto Y, Takahashi T, Saito T, Eguchi H, Doki Y. Long-Term Effects of Oral Nutritional Supplements After Gastrectomy for Gastric Cancer: A Survival Analysis from a Multicenter, Open-Label, Randomized Controlled Trial. Ann Surg Oncol 2024; 31:6909-6917. [PMID: 38985228 DOI: 10.1245/s10434-024-15667-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Weight loss (WL) after gastrectomy for gastric cancer is associated with both decreased compliance with adjuvant chemotherapy and impaired survival. This study examined the effects of administering oral nutritional supplements (ONS) for 3 months after gastrectomy in terms of compliance with adjuvant chemotherapy and survival outcomes. METHODS This large-scale, multicenter, open-label, randomized controlled trial enrolled 1,003 gastric cancer patients undergoing curative gastrectomy. Patients were assigned to the control group (n = 503) or ONS group (n = 500). In the ONS group, 400 kcal/day of ONS was recommended in addition to a regular diet for 3 months after gastrectomy. Compliance with adjuvant chemotherapy and survival outcomes were compared between the two groups. RESULTS Compared with the control group, the ONS group showed significantly decreased WL at 3 months after gastrectomy (8.6 ± 6.1 vs. 7.2 ± 5.7%, respectively, P = 0.0004). The control and ONS groups did not differ regarding the induction rate of adjuvant chemotherapy (84.9 vs. 82.8%, respectively, P = 0.614) or the continuation rate at 3 months postoperatively (75.3 vs. 76.6%, respectively, P = 0.809). Oral nutritional supplements for 3 months showed no survival benefit; the 3- and 5-year overall survival (OS) rates were 91.3% and 87.6% in the control group and 89.6% and 86.4% in the ONS group, respectively, indicating no significant difference (P = 0.548). Subgroup analysis could not detect a population in which ONS administration increased OS. CONCLUSIONS Administration of ONS for 3 months after gastrectomy was not associated with increased compliance with adjuvant chemotherapy or with improved prognosis.
Collapse
Affiliation(s)
- Takeshi Omori
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Japan
| | - Kazumasa Fujitani
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Japan
| | - Ryohei Kawabata
- Department of Gastroenterological Surgery, Sakai City Medical Center, Sakai, Japan
| | - Hiroshi Imamura
- Department of Surgery Toyonaka Municipal Hospital, Toyonaka, Japan
| | - Atsushi Takeno
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Yoshitomo Yanagimoto
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
29
|
Clarke GS, Page AJ, Eldeghaidy S. The gut-brain axis in appetite, satiety, food intake, and eating behavior: Insights from animal models and human studies. Pharmacol Res Perspect 2024; 12:e70027. [PMID: 39417406 PMCID: PMC11483575 DOI: 10.1002/prp2.70027] [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: 01/16/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
The gut-brain axis plays a pivotal role in the finely tuned orchestration of food intake, where both homeostatic and hedonic processes collaboratively control our dietary decisions. This interplay involves the transmission of mechanical and chemical signals from the gastrointestinal tract to the appetite centers in the brain, conveying information on meal arrival, quantity, and chemical composition. These signals are processed in the brain eventually leading to the sensation of satiety and the termination of a meal. However, the regulation of food intake and appetite extends beyond the realms of pure physiological need. Hedonic mechanisms, including sensory perception (i.e., through sight, smell, and taste), habitual behaviors, and psychological factors, exert profound influences on food intake. Drawing from studies in animal models and human research, this comprehensive review summarizes the physiological mechanisms that underlie the gut-brain axis and its interplay with the reward network in the regulation of appetite and satiety. The recent advancements in neuroimaging techniques, with a focus on human studies that enable investigation of the neural mechanisms underpinning appetite regulation are discussed. Furthermore, this review explores therapeutic/pharmacological strategies that hold the potential for controlling food intake.
Collapse
Affiliation(s)
- Georgia S. Clarke
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Robinson Research InstituteThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes and Gut Health, Lifelong Health ThemeSouth Australian Health and Medical Research Institute, SAHMRIAdelaideSouth AustraliaAustralia
| | - Amanda J. Page
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes and Gut Health, Lifelong Health ThemeSouth Australian Health and Medical Research Institute, SAHMRIAdelaideSouth AustraliaAustralia
| | - Sally Eldeghaidy
- Division of Food, Nutrition and DieteticsSchool of Biosciences, University of NottinghamNottinghamUK
- Sir Peter Mansfield Imaging CentreSchool of Physics and Astronomy, University of NottinghamNottinghamUK
| |
Collapse
|
30
|
King ME, Herzing HM, McLeod KR, Klotz JL, Foote AP, Edwards JL, Harmon DL. Impact of endophyte-infected tall fescue seed consumption on endocrine changes associated with intake regulation and post-absorptive metabolism in growing steers. Domest Anim Endocrinol 2024; 89:106873. [PMID: 39032187 DOI: 10.1016/j.domaniend.2024.106873] [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: 04/02/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 07/22/2024]
Abstract
Fescue toxicosis is a syndrome occurring from the consumption of endophyte-infected tall fescue and results in substantial economic losses to the beef industry primarily from reduced growth accompanied by decreased dry matter intake (DMI); however, the associations characterizing this reduction in DMI have yet to be elucidated. The objective of this experiment was to identify endocrine changes associated with intake regulation post-consumption of endophyte-infected tall fescue seed (E+). Twelve Holstein steers were stratified by body weight and assigned to 1 of 3 treatments (n=4): 0 ppm ergovaline (ERV), 1.8 ppm ERV, or 2.7 ppm ERV. Treatments were achieved by combining differing proportions of ground E+ and non-endophyte-infected tall fescue seed. Steers were adapted to their diets for 7 d followed by a 7 d DMI collection period. Within treatment, steers were assigned to a sampling day (d 16 or d 17). Blood samples were collected every 20 min for 8 h, beginning 1 h before feeding. Intake data was analyzed using the MIXED procedure of SAS 9.4 (SAS Inst. Inc., Cary, NC) with treatment, day, and the interaction as fixed effects. Hormone and metabolite data were analyzed with the fixed effect of treatment, time, and the interaction including time as a repeated measure and orthogonal contrasts. Dry matter intake was linearly decreased with increasing ERV in the diet (P < 0.001). Insulin and leptin concentrations exhibited a quadratic effect (P = 0.018 and P = 0.005) with insulin concentrations highest for the 2.7 ppm treatment and leptin concentrations highest for the 1.8 ppm treatment. No differences were detected for active ghrelin or β-hydroxybuytrate concentrations among treatment groups. Further, steers consuming both the 1.8 and 2.7 ppm ERV treatments had lower prolactin concentrations compared to the 0 ppm treatment (quadratic, P= 0.019). Glucose concentrations had a tendency for a linear increase as ERV concentrations increased (P = 0.091). A treatment × time interaction (P = 0.002) was noted in NEFA concentrations, with the 1.8 ppm ERV treatment showing increased pre-feeding concentrations, and the 2.7 ppm ERV treatment exhibiting elevated NEFA concentrations as time post-feeding progressed. The results suggest that E+ consumption reduces intake likely through alterations in intake-related hormones and post-absorptive metabolism and contributes to our current understanding of E+ effects on intake reduction while providing avenues for future research.
Collapse
Affiliation(s)
- Mindy E King
- Department of Animal and Food Science, University of Kentucky, Lexington, KY, USA
| | - Hannah M Herzing
- Department of Animal and Food Science, University of Kentucky, Lexington, KY, USA
| | - Kyle R McLeod
- Department of Animal and Food Science, University of Kentucky, Lexington, KY, USA
| | - James L Klotz
- Forage-Animal Production Research Unit, USDA-ARS, Lexington, KY, USA
| | - Andrew P Foote
- Department of Animal and Food Science, Oklahoma State University, Stillwater, OK, USA
| | - J Lannett Edwards
- Department of Animal Science, The University of Tennessee, Knoxville, TN, USA
| | - David L Harmon
- Department of Animal and Food Science, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
31
|
Dong B, Peng Y, Wang M, Peng C, Li X. Multi-omics integrated analyses indicated that non-polysaccharides of Sijunzi decoction ameliorated spleen deficiency syndrome via regulating microbiota-gut-metabolites axis and exerted synergistic compatibility. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118276. [PMID: 38697408 DOI: 10.1016/j.jep.2024.118276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a classical traditional Chinese medicine formula to invigorating spleen and replenishing qi, Sijunzi decoction (SJZD) is composed of four herbs, which is applied to cure spleen deficiency syndrome (SDS) clinically. The non-polysaccharides (NPSs) of SJZD (SJZD_NPS) are important pharmacodynamic material basis. However, the amelioration mechanism of SJZD_NPS on SDS has not been fully elaborated. Additionally, the contribution of herbs compatibility to efficacy of this formula remains unclear. AIM OF THE STUDY The aim was to explore the underlying mechanisms of SJZD_NPS on improving SDS, and uncover the scientific connotation in SJZD compatibility. MATERIALS AND METHODS A strategy integrating incomplete formulae (called "Chai-fang" in Chinese) comparison, pharmacodynamics, gut microbiome, and metabolome was employed to reveal the role of each herb to SJZD compatibility against SDS. Additionally, the underlying mechanism harbored by SJZD_NPS was further explored through targeted metabolomics, network pharmacology, molecular docking, pseudo-sterile model, and metagenomics. RESULTS SJZD_NPS significantly alleviated diarrhea, disordered secretion of gastrointestinal hormones and neurotransmitters, damage of ileal morphology and intestinal barrier in SDS rats, which was superior to the NPSs of Chai-fang. 16S rRNA gene sequencing and metabolomics analyses revealed that SJZD_NPS effectively restored the disturbed gut microbiota community and abnormal metabolism caused by SDS, showing the most evident recovery. Moreover, SJZD_NPS recalled the levels of partial amino acids, short chain fatty acids and bile acids, which possessed strong binding affinity towards potential targets. The depletion of gut microbiota confirmed that the SDS-amelioration efficacy of SJZD_NPS is dependent on the intact gut microbiome, with the relative abundance of potential probiotics such as Lactobacillus_johnsonii and Lactobacillus_taiwanensis been enriched. CONCLUSION NPSs in SJZD can improve SDS-induced gastrointestinal-nervous system dysfunction through regulating microbiota-gut-metabolites axis, with four herbs exerting synergistic effects, which indicated the compatibility rationality of SJZD.
Collapse
Affiliation(s)
- Bangjian Dong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mengyue Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chongsheng Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaobo Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
32
|
Cheng Y, He J, Zheng P, Yu J, Pu J, Huang Z, Mao X, Luo Y, Luo J, Yan H, Wu A, Yu B, Chen D. Effects of replacing soybean meal with enzymolysis-fermentation compound protein feed on growth performance, apparent digestibility of nutrients, carcass traits, and meat quality in growing-finishing pigs. J Anim Sci Biotechnol 2024; 15:127. [PMID: 39261875 PMCID: PMC11391718 DOI: 10.1186/s40104-024-01080-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/24/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Addressing the shortage of high-quality protein resources, this study was conducted to investigate the effects of replacing soybean meal (SBM) with different levels of enzymolysis-fermentation compound protein feed (EFCP) in the diets of growing-finishing pigs, focusing on growth performance, nutrients digestibility, carcass traits, and meat quality. METHODS Sixty DLY (Duroc × Landrace × Yorkshire) pigs with an initial body weight of 42.76 ± 2.05 kg were assigned to 5 dietary treatments in a 2 × 2 + 1 factorial design. These dietary treatments included a corn-soybean meal diet (CON), untreated compound protein feed (UCP) substitution 50% (U50) and 100% SBM (U100) diets, and EFCP substitution 50% (EF50) and 100% SBM (EF100) diets. Each treatment had 6 pens (replicates) with 2 pigs per pen, and the experiment lasted 58 d, divided into phase I (1-28 d) and phase II (29-58 d). Following phase I, only the CON, U50, and EF50 groups were continued for phase II, each with 5 replicate pens. On d 59, a total of 15 pigs (1 pig/pen, 5 pens/treatment) were euthanized. RESULTS During phase I, the EF50 group had a higher average daily gain (ADG) in pigs (P < 0.05) compared to the CON group, whereas the U50 group did not have a significant difference. As the substitution ratio of UCP and EFCP increased in phase I, there was a noticeable reduction in the final body weight and ADG (P < 0.05), along with an increase in the feed-to-gain ratio (F/G) (P < 0.05). In phase II, there were no significant differences in growth performance among the treatment groups, but EF50 increased the apparent digestibility of several nutrients (including dry matter, crude protein, crude fiber, acid detergent fiber, ash, gross energy) compared to U50. The EF50 group also exhibited significantly higher serum levels of neuropeptide Y and ghrelin compared to the CON and U50 groups (P < 0.05). Moreover, the EF50 group had higher carcass weight and carcass length than those in the CON and U50 groups (P < 0.05), with no significant difference in meat quality. CONCLUSIONS The study findings suggest that replacing 50% SBM with EFCP during the growing-finishing period can improve the growth performance, nutrient digestibility, and carcass traits of pigs without compromising meat quality. This research offers valuable insights into the modification of unconventional plant protein meals and developing alternatives to SBM.
Collapse
Affiliation(s)
- Yu Cheng
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Jun He
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Ping Zheng
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Jie Yu
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Junning Pu
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Zhiqing Huang
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Xiangbing Mao
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Yuheng Luo
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Junqiu Luo
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Hui Yan
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Aimin Wu
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Bing Yu
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China.
| | - Daiwen Chen
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China.
| |
Collapse
|
33
|
Gouda M, Ganesh CB. The influence of ghrelin agonist ipamorelin acetate on the hypothalamic-pituitary-testicular axis in a cichlid fish, Oreochromis mossambicus. Anim Reprod Sci 2024; 268:107550. [PMID: 38996787 DOI: 10.1016/j.anireprosci.2024.107550] [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: 05/03/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Ghrelin, a peptide found in the brain and gut, is predicted to play a significant role in the control of various physiological systems in fish. The objective of this study was to examine the impact of ipamorelin acetate (IPA), a ghrelin agonist, on the reproductive axis of the tilapia Oreochromis mossambicus. The administration of either 5 or 30 µg of IPA for 21 days led to a significant and dose-dependent rise in food intake concomitant with a significant increase in the numbers of primary spermatocytes, secondary spermatocytes, and early spermatids compared to the control group. There was a significant rise in the number of late spermatids, as well as the areas of the lobule and lumen, in fish treated with 30 µg of IPA, compared to the control group. Moreover, there was no significant difference in the percentage of gonadotropin-releasing hormone (GnRH)-immunoreactive fibres in the hypothalamus and anterior pituitary gland across different groups. However, a significant elevation in the expression of androgen receptor protein was observed in fish treated with 30 µg of IPA. Furthermore, the concentrations of luteinizing hormone (LH) and 11-ketotestosterone (11-KT) in the serum of fish treated with either 5 or 30 µg of IPA were significantly elevated in comparison to the control group. Collectively, these findings suggest that the administration of ghrelin enhances the development of germ cells during the meiosis-I phase and that this effect might be mediated via the stimulation of 11-KT and androgen receptors at the testicular level and LH at the pituitary level in the tilapia.
Collapse
Affiliation(s)
- Mallikarjun Gouda
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India
| | - C B Ganesh
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India.
| |
Collapse
|
34
|
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
|
35
|
Barbagallo F, Bosoni D, Perone V, Cucinella L, Dealberti D, Cannarella R, Calogero AE, Nappi RE. Gene-environment interaction in functional hypothalamic amenorrhea. Front Endocrinol (Lausanne) 2024; 15:1423898. [PMID: 39268244 PMCID: PMC11390525 DOI: 10.3389/fendo.2024.1423898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Functional hypothalamic amenorrhea (FHA) is a common cause of amenorrhea and chronic anovulation in adolescent girls and young women, diagnosed after excluding other organic causes. It is commonly associated with calorie restriction, excessive physical exercise, and psychosocial stress. These stressors alter the pulsatile secretion of gonadotropin-releasing hormone, leading to a chronic condition of hypoestrogenism and significant health consequences. Recent evidence has highlighted a genetic predisposition to FHA that could explain interindividual variability in stress response. Indeed, not all women experience FHA in response to stress. Rare variants in genes associated with idiopathic hypogonadotropic hypogonadism have been identified in women with FHA, suggesting that these mutations may contribute to an increased susceptibility of women to the trigger of stress exposure. FHA appears today as a complex disease resulting from the combination of genetic predisposition, environmental factors, and epigenetic changes. Furthermore, the genetic background of FHA allows for the hypothesis of a male counterpart. Despite the paucity of data, preliminary findings indicate that an equivalent condition of FHA exists in men, warranting further investigation. This narrative review aims to summarize the recent genetic evidence contributing to the pathophysiology of FHA and to raise awareness on a possible male counterpart.
Collapse
Affiliation(s)
- Federica Barbagallo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - David Bosoni
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Valeria Perone
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| | - Laura Cucinella
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| | - Davide Dealberti
- Department of Obstetrics and Gynecology, Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rossella E Nappi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS San Matteo Foundation, Pavia, Italy
| |
Collapse
|
36
|
Ma Y, Yan Q, Wang P, Guo W, Yu L. Therapeutic potential of ghrelin/GOAT/GHSR system in gastrointestinal disorders. Front Nutr 2024; 11:1422431. [PMID: 39246401 PMCID: PMC11380557 DOI: 10.3389/fnut.2024.1422431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Ghrelin, a peptide primarily secreted in the stomach, acts via the growth hormone secretagogue receptor (GHSR). It regulates several physiological processes, such as feeding behavior, energy homeostasis, glucose and lipid metabolism, cardiovascular function, bone formation, stress response, and learning. GHSR exhibits significant expression within the central nervous system. However, numerous murine studies indicate that ghrelin is limited in its ability to enter the brain from the bloodstream and is primarily confined to specific regions, such as arcuate nucleus (ARC) and median eminence (ME). Nevertheless, the central ghrelin system plays an essential role in regulating feeding behavior. Furthermore, the role of vagal afferent fibers in regulating the functions of ghrelin remains a major topic of discussion among researchers. In recent times, numerous studies have elucidated the substantial therapeutic potential of ghrelin in most gastrointestinal (GI) diseases. This has led to the development of numerous pharmaceutical agents that target the ghrelin system, some of which are currently under examination in clinical trials. Furthermore, ghrelin is speculated to serve as a promising biomarker for GI tumors, which indicates its potential use in tumor grade and stage evaluation. This review presents a summary of recent findings in research conducted on both animals and humans, highlighting the therapeutic properties of ghrelin system in GI disorders.
Collapse
Affiliation(s)
- Yunxiao Ma
- Department of Endocrinology and Metabolism of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qihui Yan
- Department of Endocrinology and Metabolism of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ping Wang
- Department of Otolaryngology-Head and Neck Surgery of First Hospital of Jilin University, Jilin University, Changchun, China
| | - Weiying Guo
- Department of Endocrinology and Metabolism of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lu Yu
- Department of Endocrinology and Metabolism of First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
37
|
Casado S, Varela-Miguéns M, de Oliveira Diz T, Quintela-Vilariño C, Nogueiras R, Diéguez C, Tovar S. The effects of ghrelin and LEAP-2 in energy homeostasis are modulated by thermoneutrality, high-fat diet and aging. J Endocrinol Invest 2024; 47:2061-2074. [PMID: 38337094 PMCID: PMC11266414 DOI: 10.1007/s40618-024-02307-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
PURPOSE Liver-expressed antimicrobial peptide 2 (LEAP-2) has been recently identified as the endogenous non-competitive allosteric antagonist of the growth hormone secretagogue receptor 1a (GHSR1a). In rodents, LEAP-2 blunts ghrelin-induced feeding and its plasma levels are modulated in response to nutritional status, being decreased upon fasting and increased in high-fat diet (HFD) fed mice. Clinical data support the regulation of circulating LEAP-2 by nutrient availability in humans. In this work, our primary objective was to examine the chronic effects of ghrelin and LEAP-2 administration on food intake, adiposity, and energy expenditure in young mice subjected to standard and HFD at both room temperature and at thermoneutrality. Furthermore, we aimed to assess the impact of these two hormones on aging mice. RESULTS Our results indicate that LEAP-2 produces a significant decrease of body weight and adiposity, an increase in energy expenditure, and activation of the thermogenic program in white and brown adipose tissue depots. However, this effect is not maintained under HFD or under thermoneutral conditions and is only partially observed in aging mice. CONCLUSION In summary our studies describe the central effects of LEAP-2 within distinct experimental contexts, and contribute to the comprehension of LEAP-2's role in energy metabolism.
Collapse
Affiliation(s)
- S Casado
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - M Varela-Miguéns
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - T de Oliveira Diz
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - C Quintela-Vilariño
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
| | - R Nogueiras
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain
| | - C Diéguez
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain.
| | - S Tovar
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), 15782, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029, Madrid, Spain.
| |
Collapse
|
38
|
Castro G, Mendes NF, Weissmann L, Quaresma PGF, Saad MJA, Prada PO. Multiple metabolic signals in the CeA regulate feeding: The role of AMPK. Mol Cell Endocrinol 2024; 589:112232. [PMID: 38604549 DOI: 10.1016/j.mce.2024.112232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND The central nucleus of the amygdala (CeA) is part of the dopaminergic reward system and controls energy balance. Recently, a cluster of neurons was identified as responsive to the orexigenic effect of ghrelin and fasting. However, the signaling pathway by which ghrelin and fasting induce feeding is unknown. AMP-activated protein kinase (AMPK) is a cellular energy sensor, and its Thr172 phosphorylation (AMPKThr172) in the mediobasal hypothalamus regulates food intake. However, whether the expression and activation of AMPK in CeA could be one of the intracellular signaling activated in response to ghrelin and fasting eliciting food intake is unknown. AIM To evaluate the activation of AMPK into CeA in response to ghrelin, fasting, and 2-deoxy-D-glucose (2DG) and whether feeding accompanied these changes. In addition, to investigate whether the inhibition of AMPK into CeA could decrease food intake. METHODS On a chow diet, eight-week-old Wistar male rats were stereotaxically implanted with a cannula in the CeA to inject several modulators of AMPKα1/2Thr172 phosphorylation, and we performed physiological and molecular assays. KEY FINDINGS Fasting increased, and refeeding reduced AMPKThr172 in the CeA. Intra-CeA glucose injection decreased feeding, whereas injection of 2DG, a glucoprivation inductor, in the CeA, increased food intake and blood glucose, despite faint increases in AMPKThr172. Intra-CeA ghrelin injection increased food intake and AMPKThr172. To further confirm the role of AMPK in the CeA, chronic injection of Melanotan II (MTII) in CeA reduced body mass and food intake over seven days together with a slight decrease in AMPKThr172. SIGNIFICANCE Our findings identified that AMPK might be part of the signaling machinery in the CeA, which responds to nutrients and hormones contributing to feeding control. The results can contribute to understanding the pathophysiological mechanisms of altered feeding behavior/consumption, such as binge eating of caloric-dense, palatable food.
Collapse
Affiliation(s)
- Gisele Castro
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Natália Ferreira Mendes
- Department of Translational Medicine (Section of Pharmacology), School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Laís Weissmann
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Mario Jose Abdalla Saad
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Patricia Oliveira Prada
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil; School of Applied Sciences, State University of Campinas (UNICAMP), Limeira, SP, Brazil; Biology Institute, State University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
39
|
Wang J, O'Reilly M, Cooper IA, Chehrehasa F, Moody H, Beecher K. Mapping GABAergic projections that mediate feeding. Neurosci Biobehav Rev 2024; 163:105743. [PMID: 38821151 DOI: 10.1016/j.neubiorev.2024.105743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Neuroscience offers important insights into the pathogenesis and treatment of obesity by investigating neural circuits underpinning appetite and feeding. Gamma-aminobutyric acid (GABA), one of the most abundant neurotransmitters in the brain, and its associated receptors represent an array of pharmacologically targetable mediators of appetite signalling. Targeting the GABAergic system is therefore an increasingly investigated approach to obesity treatment. However, the many GABAergic projections that control feeding have yet to be collectively analysed. This review provides a comprehensive analysis of the relationship between GABAergic signalling and appetite by examining both foundational studies and the results of newly emerging chemogenetic/optogenetic experiments. A current snapshot of these efforts to map GABAergic projections influencing appetite is provided, and potential avenues for further investigation are provided.
Collapse
Affiliation(s)
- Joshua Wang
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia.
| | - Max O'Reilly
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| | | | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Hayley Moody
- Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Kate Beecher
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| |
Collapse
|
40
|
Suwa Y, Kunimatsu J, Kamata A, Matsumoto M, Yamada H. A Method for Evaluating Hunger and Thirst in Monkeys by Measuring Blood Ghrelin and Osmolality Levels. eNeuro 2024; 11:ENEURO.0481-23.2024. [PMID: 39013584 PMCID: PMC11361293 DOI: 10.1523/eneuro.0481-23.2024] [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: 11/17/2023] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Hunger and thirst drive animals' consumption behavior and regulate their decision-making concerning rewards. We previously assessed the thirst states of monkeys by measuring blood osmolality under controlled water access and examined how these thirst states influenced their risk-taking behavior in decisions involving fluid rewards. However, hunger assessment in monkeys remains poorly performed. Moreover, the lack of precise measures for hunger states leads to another issue regarding how hunger and thirst states interact with each other in each individual. Thus, when controlling food access to motivate performance, it remains unclear how these two physiological needs are satisfied in captive monkeys. Here, we measured blood ghrelin and osmolality levels to respectively assess hunger and thirst in four captive macaques. Using an enzyme-linked immunosorbent assay, we identified that the levels of blood ghrelin, a widely measured hunger-related peptide hormone in humans, were high after 20 h of no food access (with ad libitum water). This reflects a typical controlled food access condition. One hour after consuming a regular dry meal, the blood ghrelin levels in three out of four monkeys decreased to within their baseline range. Additionally, blood osmolality measured from the same blood sample, the standard hematological index of hydration status, increased after consuming the regular dry meal with no water access. Thus, ghrelin and osmolality may reflect the physiological states of individual monkeys regarding hunger and thirst, suggesting that these indices can be used as tools for monitoring hunger and thirst levels that mediate an animal's decision to consume rewards.
Collapse
Affiliation(s)
- Yuki Suwa
- Academic Service Office for the Medical Science Area, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Jun Kunimatsu
- Division of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Akua Kamata
- Medical Sciences, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Masayuki Matsumoto
- Division of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Hiroshi Yamada
- Division of Biomedical Science, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
41
|
Kasim RH, Chillon TS, Eleftheriadou AM, Rijntjes E, Minich WB, Zechmann S, Schomburg L. Detection of natural autoimmunity to ghrelin in diabetes mellitus. FRONTIERS IN MEDICAL TECHNOLOGY 2024; 6:1407409. [PMID: 39070294 PMCID: PMC11272539 DOI: 10.3389/fmedt.2024.1407409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
Objective Ghrelin is an orexigenic peptide that becomes post-translationally modified. Natural autoantibodies to ghrelin (ghrelin-aAb) have been described in healthy subjects, in eating disorders and rheumatic diseases, with potential clinical relevance. Despite these important reports, the data base on the prevalence and physiological role is small and technical approaches for assessing ghrelin-aAb are few, encouraging respective research for improving knowledge on the potential endocrine significance. Methods A novel immunoprecipitation assay was generated based on a fusion protein of human ghrelin with a reporter gene. Assay quality was verified with commercial antibodies. Assay characteristics and matrix effects were determined, including stability of natural ghrelin-aAb to freezing, signal linearity in dilution experiments, and comparison of different matrices. Three groups of serum samples were analyzed for ghrelin-aAb, comprising commercial sera from healthy subjects and patients with type 1 or type 2 diabetes mellitus. Results The newly generated ghrelin-aAb assay proved sensitive, robust and reliable over a broad concentration range. Results from serum and plasma differed slightly. The signals from serum remained stable towards freezing and thawing, and in dilution experiments. Applying a mathematical criterion for outliers (P75 + 1.5-times IQR), an average prevalence of 11%-12% of positive samples was identified in the different human cohorts, with no significant sex-or disease-related difference. General significance A novel diagnostic autoantibody assay detected ghrelin-aAb with a similar prevalence in diabetic patients and controls, suggesting that autoimmunity to ghrelin plays little role in diabetes mellitus, but may be of relevance in other diseases where ghrelin signaling is essential.
Collapse
Affiliation(s)
- Rega H. Kasim
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Division of Diabetes and Endocrinology, GZO Zurich Regional Health Center, Wetzikon, Switzerland
| | - Thilo Samson Chillon
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | | | - Eddy Rijntjes
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Waldemar B. Minich
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Zechmann
- Division of Diabetes and Endocrinology, GZO Zurich Regional Health Center, Wetzikon, Switzerland
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
42
|
Mohr SM, Dai Pra R, Platt MP, Feketa VV, Shanabrough M, Varela L, Kristant A, Cao H, Merriman DK, Horvath TL, Bagriantsev SN, Gracheva EO. Hypothalamic hormone deficiency enables physiological anorexia in ground squirrels during hibernation. Nat Commun 2024; 15:5803. [PMID: 38987241 PMCID: PMC11236985 DOI: 10.1038/s41467-024-49996-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024] Open
Abstract
Mammalian hibernators survive prolonged periods of cold and resource scarcity by temporarily modulating normal physiological functions, but the mechanisms underlying these adaptations are poorly understood. The hibernation cycle of thirteen-lined ground squirrels (Ictidomys tridecemlineatus) lasts for 5-7 months and comprises weeks of hypometabolic, hypothermic torpor interspersed with 24-48-h periods of an active-like interbout arousal (IBA) state. We show that ground squirrels, who endure the entire hibernation season without food, have negligible hunger during IBAs. These squirrels exhibit reversible inhibition of the hypothalamic feeding center, such that hypothalamic arcuate nucleus neurons exhibit reduced sensitivity to the orexigenic and anorexigenic effects of ghrelin and leptin, respectively. However, hypothalamic infusion of thyroid hormone during an IBA is sufficient to rescue hibernation anorexia. Our results reveal that thyroid hormone deficiency underlies hibernation anorexia and demonstrate the functional flexibility of the hypothalamic feeding center.
Collapse
Affiliation(s)
- Sarah M Mohr
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Rafael Dai Pra
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Maryann P Platt
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Viktor V Feketa
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Marya Shanabrough
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar Street, New Haven, CT, 06510, USA
| | - Luis Varela
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar Street, New Haven, CT, 06510, USA
- Laboratory of Glia-Neuron Interactions in the Control of Hunger. Achucarro_Basque Center for Neuroscience, 48940, Leioa, Vizcaya, Spain
- IKERBASQUE, Basque Foundation for Science, 48009, Bilbao, Vizcaya, Spain
| | - Ashley Kristant
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar Street, New Haven, CT, 06510, USA
| | - Haoran Cao
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Dana K Merriman
- Department of Biology, University of Wisconsin-Oshkosh, 800 Algoma Boulevard, Oshkosh, WI, 54901, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar Street, New Haven, CT, 06510, USA
- Laboratory of Glia-Neuron Interactions in the Control of Hunger. Achucarro_Basque Center for Neuroscience, 48940, Leioa, Vizcaya, Spain
- IKERBASQUE, Basque Foundation for Science, 48009, Bilbao, Vizcaya, Spain
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.
- Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.
| |
Collapse
|
43
|
ISHIMARU M, TSUCHIYA T, ENDO Y, MATSUI A, OHMURA H, MURASE H, KOROSUE K, SATO F, TAYA K. Effects of different winter paddock management of Thoroughbred weanlings and yearlings in the cold region of Japan on physiological function, endocrine function and growth. J Vet Med Sci 2024; 86:756-768. [PMID: 38777756 PMCID: PMC11251821 DOI: 10.1292/jvms.24-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Effects of different winter paddock management of Thoroughbred weanlings and yearlings in Hokkaido, Japan, which is extremely cold in winter, on physiological function, endocrine function and growth were investigated. They were divided into two groups; those kept outdoors for 22 hr in the paddock (22hr group) and those kept outdoors for 7 hr in daytime with walking exercise for 1 hr using the horse-walker (7hr+W group), and the changes in daily distance travelled, body temperature (BT), heart rate (HR), HR variability (HRV), endocrine function and growth parameters were compared between the two groups from November at the year of birth to January at 1 year of age. The 7hr+W group could travel almost the same distance as the 22hr group by using the horse-walker. The 22hr group had a lower rate of increase in body weight than the 7hr+W group in January. In addition, lower in BT and HR were observed, and HRV analysis showed an increase in high frequency power spectral density, indicating that parasympathetic nervous activity was dominant. And also, changes in circulating cortisol and thyroxine were not observed despite cold environment. On the other hand, the 7hr+W group had higher prolactin and insulin like growth factor than the 22hr group in January, and cortisol and thyroxine were also increased. Physiological and endocrinological findings from the present study indicate that the management of the 7hr+W group is effective in promoting growth and maintaining metabolism during the winter season.
Collapse
Affiliation(s)
- Mutsuki ISHIMARU
- International Department, Japan Racing Association, Tokyo, Japan
| | | | - Yoshiro ENDO
- Hidaka Training and Research Center, Japan Racing Association, Hokkaido, Japan
| | - Akira MATSUI
- Hidaka Training and Research Center, Japan Racing Association, Hokkaido, Japan
| | - Hajime OHMURA
- Hidaka Training and Research Center, Japan Racing Association, Hokkaido, Japan
| | | | - Kenji KOROSUE
- Equine Department, Japan Racing Association, Tokyo, Japan
| | - Fumio SATO
- Japan Farriery Association, Tokyo, Japan
| | - Kazuyoshi TAYA
- Laboratory of Veterinary Physiology, Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology,
Tokyo, Japan
| |
Collapse
|
44
|
Smith A, MacAulay B, Scheufen J, Hudak A, Abizaid A. Chronic Social Defeat Stress Increases Brain Permeability to Ghrelin in Male Mice. eNeuro 2024; 11:ENEURO.0093-24.2024. [PMID: 38937108 PMCID: PMC11253241 DOI: 10.1523/eneuro.0093-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/05/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024] Open
Abstract
Ghrelin is a stomach-derived hormone that increases feeding and is elevated in response to chronic psychosocial stressors. The effects of ghrelin on feeding are mediated by the binding of ghrelin to the growth hormone secretagogue receptor (GHSR), a receptor located in hypothalamic and extrahypothalamic regions important for regulating food intake and metabolic rate. The ability of ghrelin to enter the brain, however, seems to be restricted to circumventricular organs like the median eminence and the brainstem area postrema, whereas ghrelin does not readily enter other GHSR-expressing regions like the ventral tegmental area (VTA). Interestingly, social stressors result in increased blood-brain barrier permeability, and this could therefore facilitate the entry of ghrelin into the brain. To investigate this, we exposed mice to social defeat stress for 21 d and then peripherally injected a Cy5-labelled biologically active ghrelin analog. The results demonstrate that chronically stressed mice exhibit higher Cy5-ghrelin fluorescence in several hypothalamic regions in addition to the ARC, including the hippocampus and midbrain. Furthermore, Cy5-ghrelin injections resulted in increased FOS expression in regions associated with the reward system in chronically stressed mice. Further histologic analyses identified a reduction in the branching of hypothalamic astrocytes in the ARC-median eminence junction, suggesting increased blood-brain barrier permeability. These data support the hypothesis that during metabolically challenging conditions like chronic stress, ghrelin may be more able to cross the blood-brain barrier and diffuse throughout the brain to target GHSR-expressing brain regions away from circumventricular organs.
Collapse
Affiliation(s)
- Andrea Smith
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Brenna MacAulay
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Jessica Scheufen
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Abagael Hudak
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S5B6, Canada
| |
Collapse
|
45
|
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
|
46
|
Ngernsoungnern P, Rungsawang P, Janthaweera A, Duangsuwan P, Saowakon N, Sritangos P, Ngernsoungnern A. Ultrastructural study of neuronal cells and localization of ghrelin-like peptide and its receptor in the ganglia of the golden apple snail (Pomacea canaliculata). Tissue Cell 2024; 88:102348. [PMID: 38493758 DOI: 10.1016/j.tice.2024.102348] [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: 10/29/2023] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Pomacea canaliculata is an invasive snail species causing major problems in agriculture. The snail biology was then investigated. The main objective of the present study was to investigate the nervous system of the snail. The nervous system comprises pairs of cerebral, buccal, pedal, pleural, parietal ganglia and an unpaired visceral ganglion. Most neurons were concentrated at the periphery of the ganglia. The neurons were classified into four types: NR1, NR2, NR3, and NR4. The percentages of the NR3 and NR4 in the pleural and pedal ganglia were significantly higher than those of other ganglia. Ultrastructural study revealed that nuclei of all neuronal types exhibited mostly euchromatins. Many organelles including ribosomes and endoplasmic reticulum were found in their cytoplasm. However, various mitochondria were found in the NR2 and NR3. The immunohistochemistry revealed immunoreactivity of ghrelin-like peptide in the neurons of the cerebral, pleural and pedal ganglia. However, immunoreactivity of GHS-R1a-like peptide existed only in the neurons of the pleural and pedal ganglia. The present study is the first to demonstrate the existence of ghrelin-like peptide and its receptor in P. canaliculata nervous system.
Collapse
Affiliation(s)
- Piyada Ngernsoungnern
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Piyachat Rungsawang
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | | | - Pornsawan Duangsuwan
- Anatomy Program, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Naruwan Saowakon
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Pishyaporn Sritangos
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Apichart Ngernsoungnern
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
47
|
Hong SH, Choi KM. Gut hormones and appetite regulation. Curr Opin Endocrinol Diabetes Obes 2024; 31:115-121. [PMID: 38511400 DOI: 10.1097/med.0000000000000859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
PURPOSE OF REVIEW Various gut hormones interact with the brain through delicate communication, thereby influencing appetite and subsequent changes in body weight. This review summarizes the effects of gut hormones on appetite, with a focus on recent research. RECENT FINDINGS Ghrelin is known as an orexigenic hormone, whereas glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin (CCK), postprandial peptide YY (PYY), and oxyntomodulin (OXM) are known as anorexigenic hormones. Recent human studies have revealed that gut hormones act differently in various systems, including adipose tissue, beyond appetite and energy intake, and even involve in high-order thinking. Environmental factors including meal schedule, food contents and quality, type of exercise, and sleep deprivation also play a role in the influence of gut hormone on appetite, weight change, and obesity. Recently published studies have shown that retatrutide, a triple-agonist of GLP-1, GIP, and glucagon receptor, and orforglipron, a GLP-1 receptor partial agonist, are effective in weight loss and improving various metabolic parameters associated with obesity. SUMMARY Various gut hormones influence appetite, and several drugs targeting these receptors have been reported to exert positive effects on weight loss in humans. Given that diverse dietary and environmental factors affect the actions of gut hormones and appetite, there is a need for integrated and largescale long-term studies in this field.
Collapse
Affiliation(s)
- So-Hyeon Hong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
48
|
Lékó AH, Gregory-Flores A, Marchette RCN, Gomez JL, Vendruscolo JCM, Repunte-Canonigo V, Choung V, Deschaine SL, Whiting KE, Jackson SN, Cornejo MP, Perello M, You ZB, Eckhaus M, Rasineni K, Janda KD, Zorman B, Sumazin P, Koob GF, Michaelides M, Sanna PP, Vendruscolo LF, Leggio L. Genetic or pharmacological GHSR blockade has sexually dimorphic effects in rodents on a high-fat diet. Commun Biol 2024; 7:632. [PMID: 38796563 PMCID: PMC11127961 DOI: 10.1038/s42003-024-06303-5] [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: 09/29/2023] [Accepted: 05/08/2024] [Indexed: 05/28/2024] Open
Abstract
The stomach-derived hormone ghrelin regulates essential physiological functions. The ghrelin receptor (GHSR) has ligand-independent actions; therefore, GHSR gene deletion may be a reasonable approach to investigate the role of this system in feeding behaviors and diet-induced obesity (DIO). Here, we investigate the effects of a long-term (12-month) high-fat (HFD) versus regular diet on obesity-related measures in global GHSR-KO and wild-type (WT) Wistar male and female rats. Our main findings are that the GHSR gene deletion protects against DIO and decreases food intake during HFD in male but not in female rats. GHSR gene deletion increases thermogenesis and brain glucose uptake in male rats and modifies the effects of HFD on brain glucose metabolism in a sex-specific manner, as assessed with small animal positron emission tomography. We use RNA-sequencing to show that GHSR-KO rats have upregulated expression of genes responsible for fat oxidation in brown adipose tissue. Central administration of a novel GHSR inverse agonist, PF-5190457, attenuates ghrelin-induced food intake, but only in male, not in female mice. HFD-induced binge-like eating is reduced by inverse agonism in both sexes. Our results support GHSR as a promising target for new pharmacotherapies for obesity.
Collapse
Affiliation(s)
- András H Lékó
- 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, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Gregory-Flores
- 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, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Renata C N Marchette
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Vicky Choung
- 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, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, 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, MD, USA
| | - Kimberly E Whiting
- 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, MD, USA
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Shelley N Jackson
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Maria Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - 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 Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Eckhaus
- Pathology Service, Division of Veterinary Resources, Office of Research Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, 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
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Barry Zorman
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Pietro P Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, 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, MD, 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, Baltimore, MD, USA.
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, 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.
| |
Collapse
|
49
|
Ida T, Tominaga H, Iwamoto E, Kurogi A, Okura A, Shimada K, Kato J, Kuwano A, Ode H, Nagata S, Kitamura K, Yazawa T, Sato-Hashimoto M, Yasuda M, Miyazato M, Shiimura Y, Sato T, Kojima M. Acyl modifications in bovine, porcine, and equine ghrelins. Front Endocrinol (Lausanne) 2024; 15:1411483. [PMID: 38828411 PMCID: PMC11140078 DOI: 10.3389/fendo.2024.1411483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Ghrelin is a peptide hormone with various important physiological functions. The unique feature of ghrelin is its serine 3 acyl-modification, which is essential for ghrelin activity. The major form of ghrelin is modified with n-octanoic acid (C8:0) by ghrelin O-acyltransferase. Various acyl modifications have been reported in different species. However, the underlying mechanism by which ghrelin is modified with various fatty acids remains to be elucidated. Herein, we report the purification of bovine, porcine, and equine ghrelins. The major active form of bovine ghrelin was a 27-amino acid peptide with an n-octanoyl (C8:0) modification at Ser3. The major active form of porcine and equine ghrelin was a 28-amino acid peptide. However, porcine ghrelin was modified with n-octanol (C8:0), whereas equine ghrelin was modified with n-butanol (C4:0) at Ser3. This study indicates the existence of structural divergence in ghrelin and suggests that it is necessary to measure the minor and major forms of ghrelin to fully understand its physiology.
Collapse
Affiliation(s)
- Takanori Ida
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Hatsumi Tominaga
- Miyazaki Prefecture Industrial Technology Center, Miyazaki, Japan
| | - Eri Iwamoto
- Clinical Research Center, Kurume University Hospital, Fukuoka, Japan
| | - Akito Kurogi
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Ayaka Okura
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Kengo Shimada
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Johji Kato
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Atsutoshi Kuwano
- Equine Research Institute, Japan Racing Association, Tochigi, Japan
| | - Hirotaka Ode
- Racehorse Clinic, Ritto Training Center, Japan Racing Association, Shiga, Japan
| | - Sayaka Nagata
- Department of Food Science and Technology, Faculty of Health and Nutrition, Minami Kyushu University, Miyazaki, Japan
| | - Kazuo Kitamura
- Department of Projects Research, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Hokkaido, Japan
| | - Miho Sato-Hashimoto
- Department of Animal Pharmaceutical Science, School of Pharmaceutical Sciences, Kyusyu University of Medical Science, Miyazaki, Japan
| | - Masahiro Yasuda
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Mikiya Miyazato
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Yuki Shiimura
- Molecular Genetics, Institute of Life Sciences, Kurume University, Fukuoka, Japan
| | - Takahiro Sato
- Molecular Genetics, Institute of Life Sciences, Kurume University, Fukuoka, Japan
| | - Masayasu Kojima
- Molecular Genetics, Institute of Life Sciences, Kurume University, Fukuoka, Japan
| |
Collapse
|
50
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|