1
|
Igual-Gil C, Bishop CA, Jähnert M, Johann K, Coleman V, Baum V, Kruse M, Pfeiffer AFH, Pivovarova-Ramich O, Ost M, Kleinert M, Klaus S. GDF15 is required for maintaining subcutaneous adipose tissue lipid metabolic signature. Sci Rep 2024; 14:26989. [PMID: 39505926 DOI: 10.1038/s41598-024-77448-w] [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: 07/12/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
Recent research has identified growth differentiation factor 15 (GDF15) as a crucial factor in various physiological and pathological processes, particularly in energy balance regulation. While the role of GDF15 in modulating energy metabolism through hindbrain GDNF family receptor alpha-like (GFRAL) signaling has been extensively studied, emerging evidence suggests direct peripheral metabolic actions of GDF15. Using knockout mouse models, we investigated GDF15 and GFRAL's roles in adipose tissue metabolism. Our findings indicate that C57BL/6/129/SvJ Gdf15-KO mice exhibit impaired expression of de novo lipogenesis enzymes in subcutaneous adipose tissue (sWAT). In contrast, C57BL/6J Gfral-KO mice showed no impairments compared to wild-type (WT) littermates. RNA-Seq analysis of sWAT in Gdf15-KO mice revealed a broad downregulation of genes involved in lipid metabolism. Importantly, our study uncovered sex-specific effects, with females being more affected by GDF15 loss than males. Additionally, we observed a fasting-induced upregulation of GDF15 gene expression in sWAT of both mice and humans, reinforcing this factor's role in adipose tissue lipid metabolism. In conclusion, our research highlights an essential role for GDF15 in sWAT lipid metabolic homeostasis. These insights enhance our understanding of GDF15's functions in adipose tissue physiology and underscore its potential as a therapeutic target for metabolic disorders.
Collapse
Affiliation(s)
- Carla Igual-Gil
- Department of Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
| | - Christopher A Bishop
- Department of Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Kornelia Johann
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
| | - Verena Coleman
- Department of Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
| | - Vanessa Baum
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
| | - Michael Kruse
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Andreas F H Pfeiffer
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Olga Pivovarova-Ramich
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Molecular Metabolism and Precision Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Mario Ost
- Department of Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Maximilian Kleinert
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany
| | - Susanne Klaus
- Department of Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany.
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14458, Nuthetal, Germany.
| |
Collapse
|
2
|
Wang Q, Farhadipour M, Thijs T, Ruilova Sosoranga E, Van der Schueren B, Ceulemans LJ, Deleus E, Lannoo M, Tack J, Depoortere I. Bitter-tasting drugs tune GDF15 and GLP-1 expression via bitter taste or motilin receptors in the intestine of patients with obesity. Mol Metab 2024; 88:102002. [PMID: 39111389 PMCID: PMC11380393 DOI: 10.1016/j.molmet.2024.102002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
OBJECTIVE Growth differentiation factor 15 (GDF15), a stress related cytokine, was recently identified as a novel satiety signal acting via the GFRAL receptor located in the hindbrain. Bitter compounds are known to induce satiety via the release of glucagon-like peptide 1 (GLP-1) through activation of bitter taste receptors (TAS2Rs, 25 subtypes) on enteroendocrine cells in the gut. This study aimed to investigate whether and how bitter compounds induce a stress response in intestinal epithelial cells to affect GDF15 expression in patients with obesity, thereby facilitating satiety signaling from the gut. METHODS The acute effect of oral intake of the bitter-containing medication Plaquenil (hydroxychloroquine sulfate) on plasma GDF15 levels was evaluated in a placebo-controlled, double-blind, randomized, two-visit crossover study in healthy volunteers. Primary crypts isolated from the jejunal mucosa from patients with obesity were stimulated with vehicle or bitter compounds, and the effect on GDF15 expression was evaluated using RT-qPCR or ELISA. Immunofluorescence colocalization studies were performed between GDF15, epithelial cell type markers and TAS2Rs. The role of TAS2Rs was tested by 1) pretreatment with a TAS2R antagonist, GIV3727; 2) determining TAS2R4/43 polymorphisms that affect taste sensitivity to TAS2R4/43 agonists. RESULTS Acute intake of hydroxychloroquine sulfate increased GDF15 plasma levels, which correlated with reduced hunger scores and plasma ghrelin levels in healthy volunteers. This effect was mimicked in primary jejunal cultures from patients with obesity. GDF15 was expressed in enteroendocrine and goblet cells with higher expression levels in patients with obesity. Various bitter-tasting compounds (medicinal, plant extracts, bacterial) either increased or decreased GDF15 expression, with some also affecting GLP-1. The effect was mediated by specific intestinal TAS2R subtypes and the unfolded protein response pathway. The bitter-induced effect on GDF15/GLP-1 expression was influenced by the existence of TAS2R4 amino acid polymorphisms and TAS2R43 deletion polymorphisms that may predict patient's therapeutic responsiveness. However, the effect of the bitter-tasting antibiotic azithromycin on GDF15 release was mediated via the motilin receptor, possibly explaining some of its aversive side effects. CONCLUSIONS Bitter chemosensory and pharmacological receptors regulate the release of GDF15 from human gut epithelial cells and represent potential targets for modulating metabolic disorders or cachexia.
Collapse
Affiliation(s)
- Qian Wang
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Mona Farhadipour
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Theo Thijs
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | | | - Bart Van der Schueren
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical and Experimental Endocrinology, University of Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Leuven Intestinal Failure and Transplantation (LIFT) Center, University Hospitals Leuven, Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Matthias Lannoo
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.
| |
Collapse
|
3
|
Gurtan AM, Khalid S, Koch C, Khan MZ, Lamarche LB, Splawski I, Dolan E, Carrion AM, Zessis R, Clement ME, Chen Z, Lindsley LD, Chiu YH, Streeper RS, Denning DP, Goldfine AB, Doyon B, Abbasi A, Harrow JL, Tsunoyama K, Asaumi M, Kou I, Shuldiner AR, Rodriguez-Flores JL, Rasheed A, Jahanzaib M, Mian MR, Liaqat MB, Raza SS, Sultana R, Jalal A, Saeed MH, Abbas S, Memon FR, Ishaq M, Dominy JE, Saleheen D. Identification and characterization of human GDF15 knockouts. Nat Metab 2024; 6:1913-1921. [PMID: 39327531 DOI: 10.1038/s42255-024-01135-3] [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] [Received: 03/29/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024]
Abstract
Growth differentiation factor 15 (GDF15) is a secreted protein that regulates food intake, body weight and stress responses in pre-clinical models1. The physiological function of GDF15 in humans remains unclear. Pharmacologically, GDF15 agonism in humans causes nausea without accompanying weight loss2, and GDF15 antagonism is being tested in clinical trials to treat cachexia and anorexia. Human genetics point to a role for GDF15 in hyperemesis gravidarum, but the safety or impact of complete GDF15 loss, particularly during pregnancy, is unknown3-7. Here we show the absence of an overt phenotype in human GDF15 loss-of-function carriers, including stop gains, frameshifts and the fully inactivating missense variant C211G3. These individuals were identified from 75,018 whole-exome/genome-sequenced participants in the Pakistan Genomic Resource8,9 and recall-by-genotype studies with family-based recruitment of variant carrier probands. We describe 8 homozygous ('knockouts') and 227 heterozygous carriers of loss-of-function alleles, including C211G. GDF15 knockouts range in age from 31 to 75 years, are fertile, have multiple children and show no consistent overt phenotypes, including metabolic dysfunction. Our data support the hypothesis that GDF15 is not required for fertility, healthy pregnancy, foetal development or survival into adulthood. These observations support the safety of therapeutics that block GDF15.
Collapse
Affiliation(s)
| | - Shareef Khalid
- Columbia University Irving Medical Center, New York, NY, USA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | | | | | | | - Igor Splawski
- Biomedical Research at Novartis, Boston, MA, USA
- Yarrow Biotechnology, New York, NY, USA
| | | | | | | | | | - Zhiping Chen
- Biomedical Research at Novartis, Boston, MA, USA
| | | | - Yu-Hsin Chiu
- Biomedical Research at Novartis, Boston, MA, USA
| | | | | | | | - Brian Doyon
- Biomedical Research at Novartis, Boston, MA, USA
- Tango Therapeutics, Boston, MA, USA
| | - Ali Abbasi
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, UK
| | - Jennifer L Harrow
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, UK
| | | | | | - Ikuyo Kou
- Astellas Pharma Inc., Ibaraki, Japan
| | - Alan R Shuldiner
- Regeneron Genetics Center, LLC, Regeneron Pharmaceuticals Inc., New York, NY, USA
| | | | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | | | | | | | | | | | - Anjum Jalal
- Punjab Institute of Cardiology, Lahore, Pakistan
| | | | - Shahid Abbas
- Faisalabad Institute of Cardiology, Faisalabad, Pakistan
| | | | | | | | - Danish Saleheen
- Columbia University Irving Medical Center, New York, NY, USA.
- Center for Non-Communicable Diseases, Karachi, Pakistan.
| |
Collapse
|
4
|
Jacobsen JM, Petersen N, Torz L, Gerstenberg MK, Pedersen K, Østergaard S, Wulff BS, Andersen B, Raun K, Christoffersen BØ, John LM, Reitman ML, Kuhre RE. Housing mice near vs. below thermoneutrality affects drug-induced weight loss but does not improve prediction of efficacy in humans. Cell Rep 2024; 43:114501. [PMID: 39067024 PMCID: PMC11380917 DOI: 10.1016/j.celrep.2024.114501] [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: 02/16/2024] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Evaluation of weight loss drugs is usually performed in diet-induced obese mice housed at ∼22°C. This is a cold stress that increases energy expenditure by ∼35% compared to thermoneutrality (∼30°C), which may overestimate drug-induced weight loss. We investigated five anti-obesity mechanisms that have been in clinical development, comparing weight loss in mice housed at 22°C vs. 30°C. Glucagon-like peptide-1 (GLP-1), human fibroblast growth factor 21 (hFGF21), and melanocortin-4 receptor (MC4R) agonist induced similar weight losses. Peptide YY elicited greater vehicle-subtracted weight loss at 30°C (7.2% vs. 1.4%), whereas growth differentiation factor 15 (GDF15) was more effective at 22°C (13% vs. 6%). Independent of ambient temperature, GLP-1 and hFGF21 prevented the reduction in metabolic rate caused by weight loss. There was no simple rule for a better prediction of human drug efficacy based on ambient temperature, but since humans live at thermoneutrality, drug testing using mice should include experiments near thermoneutrality.
Collapse
Affiliation(s)
- Julie M Jacobsen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Natalia Petersen
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Lola Torz
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Kent Pedersen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Søren Østergaard
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte S Wulff
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte Andersen
- Diabetes, Obesity and NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Kirsten Raun
- Lead Portfolio Projects, Research and Early Development, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Linu M John
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Rune E Kuhre
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark; Department of Biomedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
5
|
L'homme L, Sermikli BP, Haas JT, Fleury S, Quemener S, Guinot V, Barreby E, Esser N, Caiazzo R, Verkindt H, Legendre B, Raverdy V, Cheval L, Paquot N, Piette J, Legrand-Poels S, Aouadi M, Pattou F, Staels B, Dombrowicz D. Adipose tissue macrophage infiltration and hepatocyte stress increase GDF-15 throughout development of obesity to MASH. Nat Commun 2024; 15:7173. [PMID: 39169003 PMCID: PMC11339436 DOI: 10.1038/s41467-024-51078-2] [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: 10/19/2023] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Plasma growth differentiation factor-15 (GDF-15) levels increase with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) but the underlying mechanism remains poorly defined. Using male mouse models of obesity and MASLD, and biopsies from carefully-characterized patients regarding obesity, type 2 diabetes (T2D) and MASLD status, we identify adipose tissue (AT) as the key source of GDF-15 at onset of obesity and T2D, followed by liver during the progression towards metabolic dysfunction-associated steatohepatitis (MASH). Obesity and T2D increase GDF15 expression in AT through the accumulation of macrophages, which are the main immune cells expressing GDF15. Inactivation of Gdf15 in macrophages reduces plasma GDF-15 concentrations and exacerbates obesity in mice. During MASH development, Gdf15 expression additionally increases in hepatocytes through stress-induced TFEB and DDIT3 signaling. Together, these results demonstrate a dual contribution of AT and liver to GDF-15 production in metabolic diseases and identify potential therapeutic targets to raise endogenous GDF-15 levels.
Collapse
Affiliation(s)
- Laurent L'homme
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| | - Benan Pelin Sermikli
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sébastien Fleury
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sandrine Quemener
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Valentine Guinot
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Emelie Barreby
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Nathalie Esser
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Robert Caiazzo
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Hélène Verkindt
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Benjamin Legendre
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Violeta Raverdy
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Lydie Cheval
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
- CNRS EMR 8228-Unité Métabolisme et Physiologie Rénale, Paris, France
| | - Nicolas Paquot
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-Signal Transduction, University of Liège, Liège, Belgium
| | - Sylvie Legrand-Poels
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
| | - Myriam Aouadi
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - François Pattou
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Bart Staels
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - David Dombrowicz
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
6
|
Hoekx CA, Straat ME, Bizino MB, van Eyk HJ, Lamb HJ, Smit JWA, Jazet IM, de Jager SCA, Boon MR, Martinez‐Tellez B. Growth differentiation factor 15 is not modified after weight loss induced by liraglutide in South Asians and Europids with type 2 diabetes mellitus. Exp Physiol 2024; 109:1292-1304. [PMID: 38965822 PMCID: PMC11291866 DOI: 10.1113/ep091815] [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: 02/21/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists induce weight loss in patients with type 2 diabetes mellitus (T2DM), but the underlying mechanism is unclear. Recently, the mechanism by which metformin induces weight loss could be explained by an increase in growth differentiation factor 15 (GDF15), which suppresses appetite. Therefore, we aimed to investigate whether the GLP-1R agonist liraglutide modifies plasma GDF15 levels in patients with T2DM. GDF15 levels were measured in plasma samples obtained from Dutch Europids and Dutch South Asians with T2DM before and after 26 weeks of treatment with daily liraglutide (n = 44) or placebo (n = 50) added to standard care. At baseline, circulating GDF15 levels did not differ between South Asians and Europids with T2DM. Treatment with liraglutide, compared to placebo, decreased body weight, but did not modify plasma GDF15 levels in all patients, or when data were split by ethnicity. Also, the change in plasma GDF15 levels after treatment with liraglutide did not correlate with changes in body weight or HbA1c levels. In addition, the dose of metformin used did not correlate with baseline plasma GDF15 levels. Compared to placebo, liraglutide treatment for 26 weeks does not modify plasma GDF15 levels in Dutch Europid or South Asian patients with T2DM. Thus, the weight loss induced by liraglutide is likely explained by other mechanisms beyond the GDF15 pathway. HIGHLIGHTS: What is the central question of this study? Growth differentiation factor 15 (GDF15) suppresses appetite and is increased by metformin: does the GLP-1R agonist liraglutide modify plasma GDF15 levels in patients with type 2 diabetes mellitus (T2DM)? What is the main finding and its importance? Plasma GDF15 levels did not differ between South Asians and Europids with T2DM and were not modified by 26 weeks of liraglutide in either ethnicity. Moreover, there was no correlation between the changes in plasma GDF15 levels and dosage of metformin administered, changes in body weight or HbA1c levels. The appetite-suppressing effect of liraglutide is likely exerted via pathways other than GDF15.
Collapse
Affiliation(s)
- Carlijn A. Hoekx
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Maaike E. Straat
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Maurice B. Bizino
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Huub J. van Eyk
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | | | - Johannes W. A. Smit
- Department of MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Ingrid M. Jazet
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Saskia C. A. de Jager
- Laboratory of Translational ImmunologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Borja Martinez‐Tellez
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Nursing Physiotherapy and Medicine, SPORT Research Group (CTS‐1024), CERNEP Research CenterUniversity of AlmeríaAlmeríaSpain
- Biomedical Research UnitTorrecárdenas University HospitalAlmeríaSpain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIGranadaSpain
| |
Collapse
|
7
|
Fiorenza M, Checa A, Sandsdal RM, Jensen SBK, Juhl CR, Noer MH, Bogh NP, Lundgren JR, Janus C, Stallknecht BM, Holst JJ, Madsbad S, Wheelock CE, Torekov SS. Weight-loss maintenance is accompanied by interconnected alterations in circulating FGF21-adiponectin-leptin and bioactive sphingolipids. Cell Rep Med 2024; 5:101629. [PMID: 38959886 PMCID: PMC11293340 DOI: 10.1016/j.xcrm.2024.101629] [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: 04/25/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
Weight loss is often followed by weight regain. Characterizing endocrine alterations accompanying weight reduction and regain may disentangle the complex biology of weight-loss maintenance. Here, we profile energy-balance-regulating metabokines and sphingolipids in adults with obesity undergoing an initial low-calorie diet-induced weight loss and a subsequent weight-loss maintenance phase with exercise, glucagon-like peptide-1 (GLP-1) analog therapy, both combined, or placebo. We show that circulating growth differentiation factor 15 (GDF15) and C16:0-C18:0 ceramides transiently increase upon initial diet-induced weight loss. Conversely, circulating fibroblast growth factor 21 (FGF21) is downregulated following weight-loss maintenance with combined exercise and GLP-1 analog therapy, coinciding with increased adiponectin, decreased leptin, and overall decrements in ceramide and sphingosine-1-phosphate levels. Subgroup analyses reveal differential alterations in FGF21-adiponectin-leptin-sphingolipids between weight maintainers and regainers. Clinically, cardiometabolic health outcomes associate with selective metabokine-sphingolipid remodeling signatures. Collectively, our findings indicate distinct FGF21, GDF15, and ceramide responses to diverse phases of weight change and suggest that weight-loss maintenance involves alterations within the metabokine-sphingolipid axis.
Collapse
Affiliation(s)
- Matteo Fiorenza
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rasmus M Sandsdal
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Simon B K Jensen
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christian R Juhl
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Mikkel H Noer
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nicolai P Bogh
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Julie R Lundgren
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Charlotte Janus
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bente M Stallknecht
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital-Amager and Hvidovre, 2650 Hvidovre, Denmark
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Signe S Torekov
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
8
|
Richter MM, Kemp IM, Heebøll S, Winther-Sørensen M, Kjeldsen SAS, Jensen NJ, Nybing JD, Linden FH, Høgh-Schmidt E, Boesen MP, Madsbad S, Schiødt FV, Nørgaard K, Schmidt S, Gluud LL, Haugaard SB, Holst JJ, Nielsen S, Rungby J, Wewer Albrechtsen NJ. Glucagon augments the secretion of FGF21 and GDF15 in MASLD by indirect mechanisms. Metabolism 2024; 156:155915. [PMID: 38631460 DOI: 10.1016/j.metabol.2024.155915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Glucagon receptor agonism is currently explored for the treatment of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). The metabolic effects of glucagon receptor agonism may in part be mediated by increases in circulating levels of Fibroblast Growth Factor 21 (FGF21) and Growth Differentiation Factor 15 (GDF15). The effect of glucagon agonism on FGF21 and GDF15 levels remains uncertain, especially in the context of elevated insulin levels commonly observed in metabolic diseases. METHODS We investigated the effect of a single bolus of glucagon and a continuous infusion of glucagon on plasma concentrations of FGF21 and GDF15 in conditions of endogenous low or high insulin levels. The studies included individuals with overweight with and without MASLD, healthy controls (CON) and individuals with type 1 diabetes (T1D). The direct effect of glucagon on FGF21 and GDF15 was evaluated using our in-house developed isolated perfused mouse liver model. RESULTS FGF21 and GDF15 correlated with plasma levels of insulin, but not glucagon, and their secretion was highly increased in MASLD compared with CON and T1D. Furthermore, FGF21 levels in individuals with overweight with or without MASLD did not increase after glucagon stimulation when insulin levels were kept constant. FGF21 and GDF15 levels were unaffected by direct stimulation with glucagon in the isolated perfused mouse liver. CONCLUSION The glucagon-induced secretion of FGF21 and GDF15 is augmented in MASLD and may depend on insulin. Thus, glucagon receptor agonism may augment its metabolic benefits in patients with MASLD through enhanced secretion of FGF21 and GDF15.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ida M Kemp
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sara Heebøll
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Marie Winther-Sørensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nicole J Jensen
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Janus D Nybing
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Frederik H Linden
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Erik Høgh-Schmidt
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Mikael P Boesen
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Frank Vinholt Schiødt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kirsten Nørgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Signe Schmidt
- Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Lise Lotte Gluud
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Gastro Unit, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
9
|
Yau K, Kuah R, Cherney DZI, Lam TKT. Obesity and the kidney: mechanistic links and therapeutic advances. Nat Rev Endocrinol 2024; 20:321-335. [PMID: 38351406 DOI: 10.1038/s41574-024-00951-7] [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] [Accepted: 01/02/2024] [Indexed: 02/19/2024]
Abstract
Obesity is strongly associated with the development of diabetes mellitus and chronic kidney disease (CKD), but there is evidence for a bidirectional relationship wherein the kidney also acts as a key regulator of body weight. In this Review, we highlight the mechanisms implicated in obesity-related CKD, and outline how the kidney might modulate feeding and body weight through a growth differentiation factor 15-dependent kidney-brain axis. The favourable effects of bariatric surgery on kidney function are discussed, and medical therapies designed for the treatment of diabetes mellitus that lower body weight and preserve kidney function independent of glycaemic lowering, including sodium-glucose cotransporter 2 inhibitors, incretin-based therapies and metformin, are also reviewed. In summary, we propose that kidney function and body weight are related in a bidirectional fashion, and that this interrelationship affects human health and disease.
Collapse
Affiliation(s)
- Kevin Yau
- Division of Nephrology, Department of Medicine, Toronto General Hospital, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Kuah
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada
| | - David Z I Cherney
- Division of Nephrology, Department of Medicine, Toronto General Hospital, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada.
| | - Tony K T Lam
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, UHN, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Asthana P, Wong HLX. Preventing obesity, insulin resistance and type 2 diabetes by targeting MT1-MMP. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167081. [PMID: 38367902 DOI: 10.1016/j.bbadis.2024.167081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Obesity is one of the predominant risk factors for type 2 diabetes. Despite all the modern advances in medicine, an effective drug treatment for obesity without overt side effects has not yet been found. The discovery of growth and differentiation factor 15 (GDF15), an appetite-regulating hormone, created hopes for the treatment of obesity. However, an insufficient understanding of the physiological regulation of GDF15 has been a major obstacle to mitigating GDF15-centric treatment of obesity. Our recent studies revealed how a series of proteolytic events predominantly mediated by membrane-type 1 matrix metalloproteinase (MT1-MMP/MMP14), a key cell-surface metalloproteinase involved in extracellular remodeling, contribute to the pathogenesis of metabolic disorders, including obesity and diabetes. The MT1-MMP-mediated cleavage of the GDNF family receptor-α-like (GFRAL), a key neuronal receptor of GDF15, controls the satiety center in the hindbrain, thereby regulating non-homeostatic appetite and bodyweight changes. Furthermore, increased activation of MT1-MMP does not only lead to increased risk of obesity, but also causes age-associated insulin resistance by cleaving Insulin Receptor in major metabolic tissues. Importantly, inhibition of MT1-MMP effectively protects against obesity and diabetes, revealing the therapeutic potential of targeting MT1-MMP for the management of metabolic disorders.
Collapse
Affiliation(s)
- Pallavi Asthana
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | | |
Collapse
|
11
|
Allard C, Miralpeix C, López-Gambero AJ, Cota D. mTORC1 in energy expenditure: consequences for obesity. Nat Rev Endocrinol 2024; 20:239-251. [PMID: 38225400 DOI: 10.1038/s41574-023-00934-0] [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] [Accepted: 11/29/2023] [Indexed: 01/17/2024]
Abstract
In eukaryotic cells, the mammalian target of rapamycin complex 1 (sometimes referred to as the mechanistic target of rapamycin complex 1; mTORC1) orchestrates cellular metabolism in response to environmental energy availability. As a result, at the organismal level, mTORC1 signalling regulates the intake, storage and use of energy by acting as a hub for the actions of nutrients and hormones, such as leptin and insulin, in different cell types. It is therefore unsurprising that deregulated mTORC1 signalling is associated with obesity. Strategies that increase energy expenditure offer therapeutic promise for the treatment of obesity. Here we review current evidence illustrating the critical role of mTORC1 signalling in the regulation of energy expenditure and adaptive thermogenesis through its various effects in neuronal circuits, adipose tissue and skeletal muscle. Understanding how mTORC1 signalling in one organ and cell type affects responses in other organs and cell types could be key to developing better, safer treatments targeting this pathway in obesity.
Collapse
Affiliation(s)
- Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | | | | | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France.
| |
Collapse
|
12
|
Richter MM, Thomsen MN, Skytte MJ, Kjeldsen SAS, Samkani A, Frystyk J, Magkos F, Holst JJ, Madsbad S, Krarup T, Haugaard SB, Wewer Albrechtsen NJ. Effect of a 6-Week Carbohydrate-Reduced High-Protein Diet on Levels of FGF21 and GDF15 in People With Type 2 Diabetes. J Endocr Soc 2024; 8:bvae008. [PMID: 38379856 PMCID: PMC10875725 DOI: 10.1210/jendso/bvae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Indexed: 02/22/2024] Open
Abstract
Context Fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) are increased in type 2 diabetes and are potential regulators of metabolism. The effect of changes in caloric intake and macronutrient composition on their circulating levels in patients with type 2 diabetes are unknown. Objective To explore the effects of a carbohydrate-reduced high-protein diet with and without a clinically significant weight loss on circulating levels of FGF21 and GDF15 in patients with type 2 diabetes. Methods We measured circulating FGF21 and GDF15 in patients with type 2 diabetes who completed 2 previously published diet interventions. Study 1 randomized 28 subjects to an isocaloric diet in a 6 + 6-week crossover trial consisting of, in random order, a carbohydrate-reduced high-protein (CRHP) or a conventional diabetes (CD) diet. Study 2 randomized 72 subjects to a 6-week hypocaloric diet aiming at a ∼6% weight loss induced by either a CRHP or a CD diet. Fasting plasma FGF21 and GDF15 were measured before and after the interventions in a subset of samples (n = 24 in study 1, n = 66 in study 2). Results Plasma levels of FGF21 were reduced by 54% in the isocaloric study (P < .05) and 18% in the hypocaloric study (P < .05) in CRHP-treated individuals only. Circulating GDF15 levels increased by 18% (P < .05) following weight loss in combination with a CRHP diet but only in those treated with metformin. Conclusion The CRHP diet significantly reduced FGF21 in people with type 2 diabetes independent of weight loss, supporting the role of FGF21 as a "nutrient sensor." Combining metformin treatment with carbohydrate restriction and weight loss may provide additional metabolic improvements due to the rise in circulating GDF15.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Mads N Thomsen
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
| | - Mads J Skytte
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Department of Forensic Medicine, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Amirsalar Samkani
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
| | - Jan Frystyk
- Department of Endocrinology, Odense University Hospital, Odense, 5000, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital—Hvidovre, Hvidovre, 2650, Denmark
| | - Thure Krarup
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| |
Collapse
|
13
|
Melson E, Ashraf U, Papamargaritis D, Davies MJ. What is the pipeline for future medications for obesity? Int J Obes (Lond) 2024:10.1038/s41366-024-01473-y. [PMID: 38302593 DOI: 10.1038/s41366-024-01473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 02/03/2024]
Abstract
Obesity is a chronic disease associated with increased risk of obesity-related complications and mortality. Our better understanding of the weight regulation mechanisms and the role of gut-brain axis on appetite has led to the development of safe and effective entero-pancreatic hormone-based treatments for obesity such as glucagon-like peptide-1 (GLP-1) receptor agonists (RA). Semaglutide 2.4 mg once weekly, a subcutaneously administered GLP-1 RA approved for obesity treatment in 2021, results in 15-17% mean weight loss (WL) with evidence of cardioprotection. Oral GLP-1 RA are also under development and early data shows similar WL efficacy to semaglutide 2.4 mg. Looking to the next generation of obesity treatments, combinations of GLP-1 with other entero-pancreatic hormones with complementary actions and/or synergistic potential (such as glucose-dependent insulinotropic polypeptide (GIP), glucagon, and amylin) are under investigation to enhance the WL and cardiometabolic benefits of GLP-1 RA. Tirzepatide, a dual GLP-1/GIP receptor agonist has been approved for glycaemic control in type 2 diabetes as well as for obesity management leading in up to 22.5% WL in phase 3 obesity trials. Other combinations of entero-pancreatic hormones including cagrisema (GLP-1/amylin RA) and the triple agonist retatrutide (GLP-1/GIP/glucagon RA) have also progressed to phase 3 trials as obesity treatments and early data suggests that may lead to even greater WL than tirzepatide. Additionally, agents with different mechanisms of action to entero-pancreatic hormones (e.g. bimagrumab) may improve the body composition during WL and are in early phase clinical trials. We are in a new era for obesity pharmacotherapy where combinations of entero-pancreatic hormones approach the WL achieved with bariatric surgery. In this review, we present the efficacy and safety data for the pipeline of obesity pharmacotherapies with a focus on entero-pancreatic hormone-based treatments and we consider the clinical implications and challenges that the new era in obesity management may bring.
Collapse
Affiliation(s)
- Eka Melson
- Diabetes Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Uzma Ashraf
- Diabetes Research Centre, University of Leicester College of Life Sciences, Leicester, UK
| | - Dimitris Papamargaritis
- Diabetes Research Centre, University of Leicester College of Life Sciences, Leicester, UK.
- Leicester Diabetes Centre, Leicester General Hospital, Leicester, LE5 4PW, UK.
- Department of Diabetes and Endocrinology, Kettering General Hospital NHS Foundation Trust, Kettering, NN16 8UZ, UK.
| | - Melanie J Davies
- Diabetes Research Centre, University of Leicester College of Life Sciences, Leicester, UK
- Leicester Diabetes Centre, Leicester General Hospital, Leicester, LE5 4PW, UK
| |
Collapse
|
14
|
Allard C, Cota D, Quarta C. Poly-Agonist Pharmacotherapies for Metabolic Diseases: Hopes and New Challenges. Drugs 2024; 84:127-148. [PMID: 38127286 DOI: 10.1007/s40265-023-01982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
The use of glucagon-like peptide-1 (GLP-1) receptor-based multi-agonists in the treatment of type 2 diabetes and obesity holds great promise for improving glycaemic control and weight management. Unimolecular dual and triple agonists targeting multiple gut hormone-related pathways are currently in clinical trials, with recent evidence supporting their efficacy. However, significant knowledge gaps remain regarding the biological mechanisms and potential adverse effects associated with these multi-target agents. The mechanisms underlying the therapeutic efficacy of GLP-1 receptor-based multi-agonists remain somewhat mysterious, and hidden threats may be associated with the use of gut hormone-based polyagonists. In this review, we provide a critical analysis of the benefits and risks associated with the use of these new drugs in the management of obesity and diabetes, while also exploring new potential applications of GLP-1-based pharmacology beyond the field of metabolic disease.
Collapse
Affiliation(s)
- Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France.
| |
Collapse
|
15
|
Zhang B, Chang JY, Lee MH, Ju SH, Yi HS, Shong M. Mitochondrial Stress and Mitokines: Therapeutic Perspectives for the Treatment of Metabolic Diseases. Diabetes Metab J 2024; 48:1-18. [PMID: 38173375 PMCID: PMC10850273 DOI: 10.4093/dmj.2023.0115] [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: 04/15/2023] [Accepted: 06/28/2023] [Indexed: 01/05/2024] Open
Abstract
Mitochondrial stress and the dysregulated mitochondrial unfolded protein response (UPRmt) are linked to various diseases, including metabolic disorders, neurodegenerative diseases, and cancer. Mitokines, signaling molecules released by mitochondrial stress response and UPRmt, are crucial mediators of inter-organ communication and influence systemic metabolic and physiological processes. In this review, we provide a comprehensive overview of mitokines, including their regulation by exercise and lifestyle interventions and their implications for various diseases. The endocrine actions of mitokines related to mitochondrial stress and adaptations are highlighted, specifically the broad functions of fibroblast growth factor 21 and growth differentiation factor 15, as well as their specific actions in regulating inter-tissue communication and metabolic homeostasis. Finally, we discuss the potential of physiological and genetic interventions to reduce the hazards associated with dysregulated mitokine signaling and preserve an equilibrium in mitochondrial stress-induced responses. This review provides valuable insights into the mechanisms underlying mitochondrial regulation of health and disease by exploring mitokine interactions and their regulation, which will facilitate the development of targeted therapies and personalized interventions to improve health outcomes and quality of life.
Collapse
Affiliation(s)
- Benyuan Zhang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Min Hee Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Sang-Hyeon Ju
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Hyon-Seung Yi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Korea
| |
Collapse
|
16
|
Fejzo M, Rocha N, Cimino I, Lockhart SM, Petry CJ, Kay RG, Burling K, Barker P, George AL, Yasara N, Premawardhena A, Gong S, Cook E, Rimmington D, Rainbow K, Withers DJ, Cortessis V, Mullin PM, MacGibbon KW, Jin E, Kam A, Campbell A, Polasek O, Tzoneva G, Gribble FM, Yeo GSH, Lam BYH, Saudek V, Hughes IA, Ong KK, Perry JRB, Sutton Cole A, Baumgarten M, Welsh P, Sattar N, Smith GCS, Charnock-Jones DS, Coll AP, Meek CL, Mettananda S, Hayward C, Mancuso N, O'Rahilly S. GDF15 linked to maternal risk of nausea and vomiting during pregnancy. Nature 2024; 625:760-767. [PMID: 38092039 PMCID: PMC10808057 DOI: 10.1038/s41586-023-06921-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 11/30/2023] [Indexed: 01/19/2024]
Abstract
GDF15, a hormone acting on the brainstem, has been implicated in the nausea and vomiting of pregnancy, including its most severe form, hyperemesis gravidarum (HG), but a full mechanistic understanding is lacking1-4. Here we report that fetal production of GDF15 and maternal sensitivity to it both contribute substantially to the risk of HG. We confirmed that higher GDF15 levels in maternal blood are associated with vomiting in pregnancy and HG. Using mass spectrometry to detect a naturally labelled GDF15 variant, we demonstrate that the vast majority of GDF15 in the maternal plasma is derived from the feto-placental unit. By studying carriers of rare and common genetic variants, we found that low levels of GDF15 in the non-pregnant state increase the risk of developing HG. Conversely, women with β-thalassaemia, a condition in which GDF15 levels are chronically high5, report very low levels of nausea and vomiting of pregnancy. In mice, the acute food intake response to a bolus of GDF15 is influenced bi-directionally by prior levels of circulating GDF15 in a manner suggesting that this system is susceptible to desensitization. Our findings support a putative causal role for fetally derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by prepregnancy exposure to the hormone, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.
Collapse
Affiliation(s)
- M Fejzo
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - N Rocha
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I Cimino
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S M Lockhart
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C J Petry
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - R G Kay
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Peptidomics and Proteomics Core Facility, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - K Burling
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Barker
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - A L George
- Peptidomics and Proteomics Core Facility, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - N Yasara
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, Sri Lanka
| | - A Premawardhena
- Adolescent and Adult Thalassaemia Care Center (University Medical Unit), North Colombo Teaching Hospital, Kadawatha, Sri Lanka
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - S Gong
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - E Cook
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - D Rimmington
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - K Rainbow
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - D J Withers
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Cortessis
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - P M Mullin
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - K W MacGibbon
- Hyperemesis Education and Research Foundation, Clackamas, OR, USA
| | - E Jin
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - A Kam
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - A Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - O Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - G Tzoneva
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - F M Gribble
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - G S H Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - B Y H Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Saudek
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I A Hughes
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - K K Ong
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - J R B Perry
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - A Sutton Cole
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - M Baumgarten
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - N Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - G C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D S Charnock-Jones
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - A P Coll
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C L Meek
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S Mettananda
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, Sri Lanka
- University Paediatrics Unit, Colombo North Teaching Hospital, Ragama, Sri Lanka
| | - C Hayward
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - N Mancuso
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Quantitative and Computational Biology, University of Southern California, California, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, California, CA, USA
| | - S O'Rahilly
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| |
Collapse
|
17
|
Lu JF, Zhu MQ, Xia B, Zhang NN, Liu XP, Liu H, Zhang RX, Xiao JY, Yang H, Zhang YQ, Li XM, Wu JW. GDF15 is a major determinant of ketogenic diet-induced weight loss. Cell Metab 2023; 35:2165-2182.e7. [PMID: 38056430 DOI: 10.1016/j.cmet.2023.11.003] [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: 01/31/2023] [Revised: 07/27/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023]
Abstract
A ketogenic diet (KD) has been promoted as an obesity management diet, yet its underlying mechanism remains elusive. Here we show that KD reduces energy intake and body weight in humans, pigs, and mice, accompanied by elevated circulating growth differentiation factor 15 (GDF15). In GDF15- or its receptor GFRAL-deficient mice, these effects of KD disappeared, demonstrating an essential role of GDF15-GFRAL signaling in KD-mediated weight loss. Gdf15 mRNA level increases in hepatocytes upon KD feeding, and knockdown of Gdf15 by AAV8 abrogated the obesity management effect of KD in mice, corroborating a hepatic origin of GDF15 production. We show that KD activates hepatic PPARγ, which directly binds to the regulatory region of Gdf15, increasing its transcription and production. Hepatic Pparγ-knockout mice show low levels of plasma GDF15 and significantly diminished obesity management effects of KD, which could be restored by either hepatic Gdf15 overexpression or recombinant GDF15 administration. Collectively, our study reveals a previously unexplored GDF15-dependent mechanism underlying KD-mediated obesity management.
Collapse
Affiliation(s)
- Jun Feng Lu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Meng Qing Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Na Na Zhang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao Peng Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Huan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Rui Xin Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jun Ying Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hui Yang
- National Health Commission (NHC) Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Ying Qi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao Miao Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Jiang Wei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
18
|
Klein AB, Ranea-Robles P, Nicolaisen TS, Gil C, Johann K, Quesada JP, Pistolevij N, Hviid KVR, Fich L, Offersen SM, Helge JW, Nielsen HS, Bakker J, Kleinert M, Clemmensen C. Cross-species comparison of pregnancy-induced GDF15. Am J Physiol Endocrinol Metab 2023; 325:E303-E309. [PMID: 37584611 DOI: 10.1152/ajpendo.00134.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023]
Abstract
Growth differentiation factor 15 (GDF15) is a stress-induced cytokine. Although the exact physiological function of GDF15 is not yet fully comprehended, the significant elevation of circulating GDF15 levels during gestation suggests a potential role for this hormone in pregnancy. This is corroborated by genetic association studies in which GDF15 and the GDF15 receptor, GDNF family receptor alpha like (GFRAL) have been linked to morning sickness and hyperemesis gravidarum (HG) in humans. Here, we studied GDF15 biology during pregnancy in mice, rats, macaques, and humans. In contrast to macaques and humans, mice and rats exhibited an underwhelming induction in plasma GDF15 levels in response to pregnancy (∼75-fold increase in macaques vs. ∼2-fold increase in rodents). The changes in circulating GDF15 levels were corroborated by the magnitude of Gdf15 mRNA and GDF15 protein expression in placentae from mice, rats, and macaques. These species-specific findings may help guide future studies focusing on GDF15 in pregnancy and on the evaluation of pharmacological strategies to interfere with GDF15-GFRAL signaling to treat severe nausea and HG.NEW & NOTEWORTHY In the present study pregnancy-induced changes in circulating growth differentiation factor 15 (GDF15) in rodents, rhesus macaques, and humans are mapped. In sum, it is demonstrated that humans and macaques exhibit a tremendous increase in placental and circulating GDF15 during pregnancy. In contrast, GDF15 is negligibly increased in pregnant mice and rats, questioning a physiological role for GDF15 in pregnancy in rodents.
Collapse
Affiliation(s)
- Anders Bue Klein
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Ranea-Robles
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Sand Nicolaisen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Cláudia Gil
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kornelia Johann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Muscle Physiology and Metabolism Group, German Institute of Human Nutrition (DIfE), Potsdam-Rehbrücke, Nuthetal, Germany
| | - Júlia Prats Quesada
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nina Pistolevij
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kathrine V R Hviid
- Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Line Fich
- Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone M Offersen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Jørn Wulff Helge
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Svarre Nielsen
- Fertility Department, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Obstetrics and Gynaecology, Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jaco Bakker
- Animal Science Department, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Maximilian Kleinert
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Muscle Physiology and Metabolism Group, German Institute of Human Nutrition (DIfE), Potsdam-Rehbrücke, Nuthetal, Germany
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Gouda MA, Hu MI, Cabanillas ME, Wu J, Meric-Bernstam F, Subbiah V. Weight gain in patients with RET aberrant cancers treated with brain penetrant RET selective inhibitors. Ann Oncol 2023; 34:946-948. [PMID: 37473872 PMCID: PMC11189099 DOI: 10.1016/j.annonc.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/22/2023] Open
Affiliation(s)
- M A Gouda
- Departments of Investigational Cancer Therapeutics
| | - M I Hu
- Departments of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston
| | - M E Cabanillas
- Departments of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston
| | - J Wu
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | | | - V Subbiah
- Departments of Investigational Cancer Therapeutics; Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, USA.
| |
Collapse
|
20
|
Alexopoulou F, Buch-Månson N, Pedersen SL, Vrang N, Fink LN, Strømgaard K. Identification of GDF15 peptide fragments inhibiting GFRAL receptor signaling. Peptides 2023; 168:171063. [PMID: 37495041 DOI: 10.1016/j.peptides.2023.171063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
Growth differentiation factor 15 (GDF15) is believed to be a major causative factor for cancer-induced cachexia. Recent elucidation of the central circuits involved in GDF15 function and its signaling through the glial cell-derived neurotrophic factor family receptor α-like (GFRAL) has prompted the interest of targeting the GDF15-GFRAL signaling for energy homeostasis and body weight regulation. Here, we applied advanced peptide technologies to identify GDF15 peptide fragments inhibiting GFRAL signaling. SPOT peptide arrays revealed binding of GDF15 C-terminal peptide fragments to the extracellular domain of GFRAL. Parallel solid-phase peptide synthesis allowed for generation of complementary GDF15 peptide libraries and their subsequent functional evaluation in cells expressing the GFRAL/RET receptor complex. We identified a series of C-terminal fragments of GDF15 inhibiting GFRAL activity in the micromolar range. These novel GFRAL peptide inhibitors could serve as valuable tools for further development of peptide therapeutics towards the treatment of cachexia and other wasting disorders.
Collapse
Affiliation(s)
- Flora Alexopoulou
- Gubra Aps, Hørsholm, DK-2970 Hørsholm, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | | | | | - Niels Vrang
- Gubra Aps, Hørsholm, DK-2970 Hørsholm, Denmark
| | | | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
21
|
Jena J, García-Peña LM, Pereira RO. The roles of FGF21 and GDF15 in mediating the mitochondrial integrated stress response. Front Endocrinol (Lausanne) 2023; 14:1264530. [PMID: 37818094 PMCID: PMC10561105 DOI: 10.3389/fendo.2023.1264530] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Various models of mitochondrial stress result in induction of the stress-responsive cytokines fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15). This is an adaptive mechanism downstream of the mitochondrial integrated stress response frequently associated with improvements in systemic metabolic health. Both FGF21 and GDF15 have been shown to modulate energy balance and glucose homeostasis, and their pharmacological administration leads to promising beneficial effects against obesity and associated metabolic diseases in pre-clinical models. Furthermore, endogenous upregulation of FGF21 and GDF15 is associated with resistance to diet-induced obesity (DIO), improved glucose homeostasis and increased insulin sensitivity. In this review, we highlight several studies on transgenic mouse models of mitochondrial stress and will compare the specific roles played by FGF21 and GDF15 on the systemic metabolic adaptations reported in these models.
Collapse
Affiliation(s)
| | | | - Renata O. Pereira
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
22
|
Sjøberg KA, Sigvardsen CM, Alvarado-Diaz A, Andersen NR, Larance M, Seeley RJ, Schjerling P, Knudsen JG, Katzilieris-Petras G, Clemmensen C, Jørgensen SB, De Bock K, Richter EA. GDF15 increases insulin action in the liver and adipose tissue via a β-adrenergic receptor-mediated mechanism. Cell Metab 2023; 35:1327-1340.e5. [PMID: 37473755 DOI: 10.1016/j.cmet.2023.06.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/10/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023]
Abstract
Growth differentiation factor 15 (GDF15) induces weight loss and increases insulin action in obese rodents. Whether and how GDF15 improves insulin action without weight loss is unknown. Obese rats were treated with GDF15 and displayed increased insulin tolerance 5 h later. Lean and obese female and male mice were treated with GDF15 on days 1, 3, and 5 without weight loss and displayed increased insulin sensitivity during a euglycemic hyperinsulinemic clamp on day 6 due to enhanced suppression of endogenous glucose production and increased glucose uptake in WAT and BAT. GDF15 also reduced glucagon levels during clamp independently of the GFRAL receptor. The insulin-sensitizing effect of GDF15 was completely abrogated in GFRAL KO mice and also by treatment with the β-adrenergic antagonist propranolol and in β1,β2-adrenergic receptor KO mice. GDF15 activation of the GFRAL receptor increases β-adrenergic signaling, in turn, improving insulin action in the liver and white and brown adipose tissue.
Collapse
Affiliation(s)
- Kim A Sjøberg
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Casper M Sigvardsen
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Abdiel Alvarado-Diaz
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Nicoline Resen Andersen
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mark Larance
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Peter Schjerling
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark; Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jakob G Knudsen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Georgios Katzilieris-Petras
- Section for Cell Biology and Physiology, Department of Biology, 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
| | - Sebastian Beck Jørgensen
- Global Drug Discovery, Obesity Research, Novo Nordisk, Maaloev, Denmark; Bio Innovation Hub Transformational Research Unit, Novo Nordisk, Boston, MA, USA
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland.
| | - Erik A Richter
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
Zheng S, Polidori D, Wang Y, Geist B, Lin‐Schmidt X, Furman JL, Nelson S, Nawrocki AR, Hinke SA. A long-acting GDF15 analog causes robust, sustained weight loss and reduction of food intake in an obese nonhuman primate model. Clin Transl Sci 2023; 16:1431-1444. [PMID: 37154518 PMCID: PMC10432867 DOI: 10.1111/cts.13543] [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: 02/07/2023] [Revised: 03/28/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023] Open
Abstract
Growth Differentiation Factor-15 (GDF15) is a circulating polypeptide linked to cellular stress and metabolic adaptation. GDF15's half-life is ~3 h and activates the glial cell line-derived neurotrophic factor family receptor alpha-like (GFRAL) receptor expressed in the area postrema. To characterize sustained GFRAL agonism on food intake (FI) and body weight (BW), we tested a half-life extended analog of GDF15 (Compound H [CpdH]) suitable for reduced dosing frequency in obese cynomolgus monkeys. Animals were chronically treated once weekly (q.w.) with CpdH or long-acting GLP-1 analog dulaglutide. Mechanism-based longitudinal exposure-response modeling characterized effects of CpdH and dulaglutide on FI and BW. The novel model accounts for both acute, exposure-dependent effects reducing FI and compensatory changes in energy expenditure (EE) and FI occurring over time with weight loss. CpdH had linear, dose-proportional pharmacokinetics (terminal half-life ~8 days) and treatment caused exposure-dependent reductions in FI and BW. The 1.6 mg/kg CpdH reduced mean FI by 57.5% at 1 week and sustained FI reductions of 31.5% from weeks 9-12, resulting in peak reduction in BW of 16 ± 5%. Dulaglutide had more modest effects on FI and peak BW loss was 3.8 ± 4.0%. Longitudinal modeling of both the FI and BW profiles suggested reductions in BW observed with both CpdH and dulaglutide were fully explained by exposure-dependent reductions in FI without increase in EE. Upon verification of the pharmacokinetic/pharmacodynamic relationship established in monkeys and humans for dulaglutide, we predicted that CpdH could reach double digit BW loss in humans. In summary, a long-acting GDF15 analog led to sustained reductions in FI in overweight monkeys and holds potential for effective clinical obesity pharmacotherapy.
Collapse
Affiliation(s)
- Songmao Zheng
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
- Present address:
AdageneSan DiegoCaliforniaUSA
| | | | - Yuanping Wang
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Brian Geist
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | | | | | | | - Simon A. Hinke
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| |
Collapse
|
24
|
Zhang M, Bagán A, Martínez D, Barroso E, Palomer X, Vázquez S, Escolano C, Vázquez-Carrera M. Design and Synthesis of AMPK Activators and GDF15 Inducers. Molecules 2023; 28:5468. [PMID: 37513338 PMCID: PMC10386161 DOI: 10.3390/molecules28145468] [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: 06/11/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Targeting growth differentiation factor 15 (GDF15) is a recent strategy for the treatment of obesity and type 2 diabetes mellitus (T2DM). Here, we designed, synthesized, and pharmacologically evaluated in vitro a novel series of AMPK activators to upregulate GDF15 levels. These compounds were structurally based on the (1-dibenzylamino-3-phenoxy)propan-2-ol structure of the orphan ubiquitin E3 ligase subunit protein Fbxo48 inhibitor, BC1618. This molecule showed a better potency than metformin, increasing GDF15 mRNA levels in human Huh-7 hepatic cells. Based on BC1618, structural modifications have been performed to create a collection of diversely substituted new molecules. Of the thirty-five new compounds evaluated, compound 21 showed a higher increase in GDF15 mRNA levels compared with BC1618. Metformin, BC1618, and compound 21 increased phosphorylated AMPK, but only 21 increased GDF15 protein levels. Overall, these findings indicate that 21 has a unique capacity to increase GDF15 protein levels in human hepatic cells compared with metformin and BC1618.
Collapse
Affiliation(s)
- Meijian Zhang
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine (IBUB), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Donna Martínez
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine (IBUB), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine (IBUB), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Santiago Vázquez
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Biomedicine (IBUB), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| |
Collapse
|
25
|
Lösch L, Stemmler A, Fischer A, Steinmetz J, Schuldt L, Hennig CL, Symmank J, Jacobs C. GDF15 Promotes the Osteogenic Cell Fate of Periodontal Ligament Fibroblasts, thus Affecting Their Mechanobiological Response. Int J Mol Sci 2023; 24:10011. [PMID: 37373159 DOI: 10.3390/ijms241210011] [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: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Periodontal ligament fibroblasts (PdLFs) exert important functions in oral tissue and bone remodeling following mechanical forces, which are specifically applied during orthodontic tooth movement (OTM). Located between the teeth and the alveolar bone, mechanical stress activates the mechanomodulatory functions of PdLFs including regulating local inflammation and activating further bone-remodeling cells. Previous studies suggested growth differentiation factor 15 (GDF15) as an important pro-inflammatory regulator during the PdLF mechanoresponse. GDF15 exerts its effects through both intracrine signaling and receptor binding, possibly even in an autocrine manner. The extent to which PdLFs are susceptible to extracellular GDF15 has not yet been investigated. Thus, our study aims to examine the influence of GDF15 exposure on the cellular properties of PdLFs and their mechanoresponse, which seems particularly relevant regarding disease- and aging-associated elevated GDF15 serum levels. Therefore, in addition to investigating potential GDF15 receptors, we analyzed its impact on the proliferation, survival, senescence, and differentiation of human PdLFs, demonstrating a pro-osteogenic effect upon long-term stimulation. Furthermore, we observed altered force-related inflammation and impaired osteoclast differentiation. Overall, our data suggest a major impact of extracellular GDF15 on PdLF differentiation and their mechanoresponse.
Collapse
Affiliation(s)
- Lukas Lösch
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany
| | - Albert Stemmler
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany
| | - Adrian Fischer
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany
| | - Julia Steinmetz
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany
| | - Lisa Schuldt
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany
| | | | - Judit Symmank
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany
| | - Collin Jacobs
- Department of Orthodontics, University Hospital Jena, Leutragraben 3, 07743 Jena, Germany
| |
Collapse
|
26
|
Fejzo M, Rocha N, Cimino I, Lockhart SM, Petry C, Kay RG, Burling K, Barker P, George AL, Yasara N, Premawardhena A, Gong S, Cook E, Rainbow K, Withers DJ, Cortessis V, Mullin PM, MacGibbon KW, Jin E, Kam A, Campbell A, Polasek O, Tzoneva G, Gribble FM, Yeo G, Lam B, Saudek V, Hughes IA, Ong KK, Perry J, Sutton Cole A, Baumgarten M, Welsh P, Sattar N, Smith G, Charnock Jones DS, Coll AP, Meek CL, Mettananda S, Hayward C, Mancuso N, O'Rahilly S. Fetally-encoded GDF15 and maternal GDF15 sensitivity are major determinants of nausea and vomiting in human pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.542661. [PMID: 37398065 PMCID: PMC10312505 DOI: 10.1101/2023.06.02.542661] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Human pregnancy is frequently accompanied by nausea and vomiting that may become severe and life-threatening, as in hyperemesis gravidarum (HG), the cause of which is unknown. Growth Differentiation Factor-15 (GDF15), a hormone known to act on the hindbrain to cause emesis, is highly expressed in the placenta and its levels in maternal blood rise rapidly in pregnancy. Variants in the maternal GDF15 gene are associated with HG. Here we report that fetal production of GDF15, and maternal sensitivity to it, both contribute substantially to the risk of HG. We found that the great majority of GDF15 in maternal circulation is derived from the feto-placental unit and that higher GDF15 levels in maternal blood are associated with vomiting and are further elevated in patients with HG. Conversely, we found that lower levels of GDF15 in the non-pregnant state predispose women to HG. A rare C211G variant in GDF15 which strongly predisposes mothers to HG, particularly when the fetus is wild-type, was found to markedly impair cellular secretion of GDF15 and associate with low circulating levels of GDF15 in the non-pregnant state. Consistent with this, two common GDF15 haplotypes which predispose to HG were associated with lower circulating levels outside pregnancy. The administration of a long-acting form of GDF15 to wild-type mice markedly reduced subsequent responses to an acute dose, establishing that desensitisation is a feature of this system. GDF15 levels are known to be highly and chronically elevated in patients with beta thalassemia. In women with this disorder, reports of symptoms of nausea or vomiting in pregnancy were strikingly diminished. Our findings support a causal role for fetal derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by pre-pregnancy exposure to GDF15, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.
Collapse
Affiliation(s)
- M Fejzo
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - N Rocha
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I Cimino
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S M Lockhart
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C Petry
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - R G Kay
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Peptidomics and Proteomics Core Facility, Level 4, Wellcome-MRC Institute of Metabolic Science, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - K Burling
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - P Barker
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - A L George
- Peptidomics and Proteomics Core Facility, Level 4, Wellcome-MRC Institute of Metabolic Science, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - N Yasara
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka
| | - A Premawardhena
- Adolescent and Adult Thalassaemia Care Center (University Medical Unit), North Colombo Teaching Hospital, Kadawatha, Sri Lanka
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - S Gong
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - E Cook
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - K Rainbow
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - D J Withers
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Cortessis
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - P M Mullin
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - K W MacGibbon
- Hyperemesis Education and Research Foundation, Clackamas, OR
| | - E Jin
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - A Kam
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - A Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - O Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - G Tzoneva
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - F M Gribble
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Gsh Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Byh Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Saudek
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I A Hughes
- Department of Paediatrics, Cambridge University Hospitals NHS Foundation Trust, University of Cambridge, Cambridge, UK
| | - K K Ong
- Department of Paediatrics, Cambridge University Hospitals NHS Foundation Trust, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jrb Perry
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - A Sutton Cole
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - M Baumgarten
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - N Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Gcs Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D S Charnock Jones
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - A P Coll
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C L Meek
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S Mettananda
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka
- University Paediatrics Unit, Colombo North Teaching Hospital, Ragama, Sri Lanka
| | - C Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU,16, UK
| | - N Mancuso
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
- Department of Quantitative and Computational Biology, University of Southern California
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California
| | - S O'Rahilly
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Jaschke NP, Rachner TD. Cancer cachexia as a blueprint for treating obesity. Trends Endocrinol Metab 2023:S1043-2760(23)00087-5. [PMID: 37173233 DOI: 10.1016/j.tem.2023.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023]
Abstract
Effective pharmacological treatments to achieve significant and sustained weight loss in obese individuals remain limited. Here, we apply a 'reverse engineering' approach to cancer cachexia, an extreme form of dysregulated energy balance resulting in net catabolism. We discuss three phenotypic features of the disease, summarize the underlying molecular checkpoints, and explore their translation to obesity research. We then provide examples for established pharmaceuticals, which follow a reverse engineering logic, and propose additional targets that may be of relevance for future studies. Finally, we argue that approaching diseases from this perspective may prove useful as a generic strategy to fuel the development of innovative therapies.
Collapse
Affiliation(s)
- Nikolai P Jaschke
- Division of Endocrinology, Department of Medicine III, Technische Universität Dresden, Dresden, Germany.
| | - Tilman D Rachner
- Division of Endocrinology, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
28
|
Davies I, Tan TMM. Design of novel therapeutics targeting the glucose-dependent insulinotropic polypeptide receptor (GIPR) to aid weight loss. Expert Opin Drug Discov 2023; 18:659-669. [PMID: 37154171 DOI: 10.1080/17460441.2023.2203911] [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: 05/10/2023]
Abstract
INTRODUCTION With obesity rates growing globally, there is a paramount need for new obesity pharmacotherapies to tackle this pandemic. AREAS COVERED This review focuses on the design of therapeutics that target the glucose-dependent insulinotropic polypeptide receptor (GIPR) to aid weight loss. The authors highlight the paradoxical observation that both GIPR agonism and antagonism appear to provide metabolic benefits when combined with glucagon-like peptide-1 receptor (GLP-1 R) agonism. The therapeutic potential of compounds that target the GIPR alongside the GLP-1 R and the glucagon receptor are discussed, and the impressive clinical findings of such compounds are reviewed. EXPERT OPINION In this area, the translation of pre-clinical findings to clinical studies appears to be particularly difficult. Well-designed physiological studies in man are required to answer the paradox highlighted above, and to support the safe future development of a combination of GLP-1 R/GIPR targeting therapies.
Collapse
Affiliation(s)
- Iona Davies
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Tricia M M Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| |
Collapse
|
29
|
Abstract
There is increasing interest in GDF15 analogs as therapeutic agents for obesity. In this issue of Cell Metabolism, Benichou et al. report the first clinical trial of such a drug in obese humans.
Collapse
Affiliation(s)
- Samuel N Breit
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney and University of New South Wales, Sydney, NSW 2010, Australia.
| | - David A Brown
- Department of Immunopathology, Institute for Clinical Pathology and Medical Research-New South Wales Health Pathology, Westmead Hospital, Sydney, NSW, Australia
| | - Vicky Wang Wei Tsai
- St. Vincent's Centre for Applied Medical Research, St. Vincent's Hospital, Sydney and University of New South Wales, Sydney, NSW 2010, Australia
| |
Collapse
|