1
|
Townsend LK, Wang D, Knuth CM, Fayyazi R, Mohammad A, Becker LJ, Tsakiridis EE, Desjardins EM, Patel Z, Valvano CM, Lu J, Payne AE, Itua O, Medak KD, Marko DM, Schertzer JD, Wright DC, Beaudette SM, Morrison KM, Carpentier AC, Blondin DP, MacPherson REK, McCall JG, Jeschke MG, Steinberg GR. GDF15 links adipose tissue lipolysis with anxiety. Nat Metab 2025:10.1038/s42255-025-01264-3. [PMID: 40234625 DOI: 10.1038/s42255-025-01264-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/06/2025] [Indexed: 04/17/2025]
Abstract
Psychological stress changes both behaviour and metabolism to protect organisms. Adrenaline is an important driver of this response. Anxiety correlates with circulating free fatty acid levels and can be alleviated by a peripherally restricted β-blocker, suggesting a peripheral signal linking metabolism with behaviour. Here we show that adrenaline, the β3 agonist CL316,243 and acute restraint stress induce growth differentiation factor 15 (GDF15) secretion in white adipose tissue of mice. Genetic inhibition of adipose triglyceride lipase or genetic deletion of β-adrenergic receptors blocks β-adrenergic-induced increases in GDF15. Increases in circulating GDF15 require lipolysis-induced free fatty acid stimulation of M2-like macrophages within white adipose tissue. Anxiety-like behaviour elicited by adrenaline or restraint stress is eliminated in mice lacking the GDF15 receptor GFRAL. These data provide molecular insights into the mechanisms linking metabolism and behaviour and suggest that inhibition of GDF15-GFRAL signalling might reduce acute anxiety.
Collapse
Affiliation(s)
- Logan K Townsend
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Carly M Knuth
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Russta Fayyazi
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ahmad Mohammad
- Department of Health Science, Brock University, St. Catherines, Ontario, Canada
| | - Léa J Becker
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Evangelia E Tsakiridis
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Eric M Desjardins
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zeel Patel
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Celina M Valvano
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Junfeng Lu
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alice E Payne
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ofure Itua
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Kyle D Medak
- Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Daniel M Marko
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - David C Wright
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shawn M Beaudette
- Department of Kinesiology, Brock University, St. Catherines, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - André C Carpentier
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Denis P Blondin
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Jordan G McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Marc G Jeschke
- David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
- Hamilton General Hospital, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Yıldırım Ayaz E, Mesci B, Üner ÖE, Kaya FN, Dincer B, İşman FK, Oğuz A. The effect of exercise on GDF-15 levels in individuals with prediabetes: A randomized controlled trial. J Diabetes Investig 2025; 16:656-669. [PMID: 39868911 PMCID: PMC11970312 DOI: 10.1111/jdi.14404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/12/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
AIMS Growth differentiation factor-15 (GDF-15) is an inflammatory cytokine that increases in prediabetes and is known for its anorexigenic effects. This study aims to evaluate the effects of a 12-week exercise program on GDF-15 in individuals with prediabetes. MATERIALS AND METHODS In this multicenter, parallel-group, randomized-controlled trial, 64 patients aged 18-60 diagnosed with prediabetes were randomized in a 1:1 ratio into the exercise group (E) and the control group (C). Additionally, 32 patients who were planned to start metformin were included in the metformin group (M). Participants in the exercise group engaged in aerobic exercise at 50-70% of their maximum heart rate for 60 min, 3 days a week. Serum GDF-15 levels were evaluated at the beginning and the end of the 12th week. RESULTS The mean age of the 91 participants who completed the study was 46.13 ± 8.52 years, and 23.1% were male. Basal GDF-15 levels were similar among the groups (E = 668.6 ± 415.1, C = 651.8 ± 352.5, M = 603.6 ± 387.2, P = 0.47). At the 12th week, GDF-15 levels were lower in the E compared to the C, while higher in the M compared to the C (E = 383.1 ± 215.6, C = 556.4 ± 285.6, M = 810.8 ± 498.0, P < 0.001). In inter-group comparisons, no significant change was observed in the C between the 0th and 12th weeks, while GDF-15 decreased in the E (P < 0.001) and increased in the M (P < 0.001). CONCLUSIONS It was determined that in individuals with prediabetes, GDF-15, which serves both as a biomarker of metabolic disorder and has a negative regulatory effect on appetite, decreased with 12 weeks of aerobic exercise and increased with metformin administration.
Collapse
Affiliation(s)
- Elif Yıldırım Ayaz
- Sultan 2. Abdülhamid Han Training and Research HospitalUniversity of Health SciencesÜsküdar, İstanbulTurkey
| | - Banu Mesci
- Göztepe Prof. Dr. Süleyman Yalçın City HospitalIstanbul Medeniyet UniversityİstanbulTurkey
| | - Özden Ezgi Üner
- Fatih Sultan Mehmet Training and Research HospitalUniversity of Health SciencesÜsküdar, İstanbulTurkey
| | | | - Berna Dincer
- Faculty of Health SciencesIstanbul Medeniyet UniversityİstanbulTurkey
| | - Ferruh Kemal İşman
- Göztepe Prof. Dr. Süleyman Yalçın City HospitalIstanbul Medeniyet UniversityİstanbulTurkey
| | - Aytekin Oğuz
- Göztepe Prof. Dr. Süleyman Yalçın City HospitalIstanbul Medeniyet UniversityİstanbulTurkey
| |
Collapse
|
3
|
Chaouki G, Parry L, Vituret C, Jousse C, Leremboure M, Bourgne C, Mosoni L, Delorme Y, Djelloul-Mazouz M, Hermet J, Averous J, Bruhat A, Combaret L, Taillandier D, Papet I, Bindels LB, Fafournoux P, Maurin AC. Pre-cachectic changes in amino acid homeostasis precede activation of eIF2α signaling in the liver at the onset of C26 cancer-induced cachexia. iScience 2025; 28:112030. [PMID: 40124481 PMCID: PMC11928868 DOI: 10.1016/j.isci.2025.112030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 11/28/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025] Open
Abstract
The sequence of events associated with cancer cachexia induction needs to be further characterized. Using the C26 mouse model, we found that prior to cachexia, cancer progression was associated with increased levels of IL-6 and growth differentiation factor 15 (GDF15), highly induced production of positive acute phase proteins (APPs) and reduced levels of most amino acids in the systemic circulation, while signal transducer and activator of transcription 3 (STAT3) signaling was induced (1) in the growing spleen, alongside activation of ribosomal protein S6 (rpS6) and alpha subunit of eukaryotic translation initiation factor-2 (eIF2α) signalings, and (2) in the liver, alongside increased positive-APP expression, decreased albumin expression, and upregulation of autophagy. At the onset of cachexia, rpS6 and eIF2α signalings were concomitantly activated in the liver, with increased expression of activating transcription factor 4 (ATF4) target genes involved in amino acid synthesis and transport, as well as autophagy. Data show that pre-cachectic (pre-Cx) alterations in protein/aa homeostasis are followed by activation of eIF2α signaling in the liver, an adaptive mechanism likely regulating protein/amino acid metabolism upon progression to cachexia.
Collapse
Affiliation(s)
- Ghita Chaouki
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Laurent Parry
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Cyrielle Vituret
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Céline Jousse
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Martin Leremboure
- Université Clermont Auvergne, Clermont Auvergne INP, CNRS, Institut de Chimie de Clermont-Ferrand (ICCF), 63000 Clermont-Ferrand, France
| | - Céline Bourgne
- Digital PCR Platform Facility of the CHU of Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Laurent Mosoni
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Yoann Delorme
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Mehdi Djelloul-Mazouz
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Julien Hermet
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Julien Averous
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Alain Bruhat
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Lydie Combaret
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Daniel Taillandier
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Isabelle Papet
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Brussels, Belgium
- Welbio Department, WEL Research Institute, Wavre, Belgium
| | - Pierre Fafournoux
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| | - Anne-Catherine Maurin
- Unité de Nutrition Humaine, INRAE, Université Clermont Auvergne, UMR 1019, F-63000 Clermont-Ferrand, France
| |
Collapse
|
4
|
Hanada S, Ishikawa K, Shirai T, Takemasa T, Nakada K. Endurance swimming exacerbates mitochondrial myopathy in mice with high mtDNA deletions. Mitochondrion 2025; 81:102010. [PMID: 39956167 DOI: 10.1016/j.mito.2025.102010] [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: 12/09/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Recent studies have reported that endurance exercise enhances mitochondrial function, facilitating discussions of its potential as a therapeutic strategy for mitochondrial diseases caused by the accumulation of mutant mitochondrial DNA (mtDNA). In this study, we assessed the effects of endurance exercise on muscle pathology in a mitochondrial disease mouse model (mito-miceΔ) that is characterized by severe clinical phenotypes owing to the predominant accumulation of mtDNA with a large-scale deletion (ΔmtDNA). Contrary to expectations that endurance exercise may enhance mitochondrial function, endurance exercise exacerbated muscle pathology in mito-miceΔ. Therefore, exercise interventions should be potentially avoided in patients with severe mitochondrial diseases.
Collapse
Affiliation(s)
- Sho Hanada
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan
| | - Kaori Ishikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.
| | - Takanaga Shirai
- Institute of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan; Research Fellow of Japan Society for Promotion Science, Chiyoda-ku, Tokyo 102-0083, Japan; Department of Human Sciences, Kanagawa University, 3-27-1 Rokkakubashi, Kanagawa-ku, Yokohama-shi, Kanagawa 221-8686, Japan
| | - Tohru Takemasa
- Institute of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
| | - Kazuto Nakada
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.
| |
Collapse
|
5
|
Sammut MJ, Thorne BR, Melling CWJ. Skeletal muscle growth to combat diabetes and obesity: the potential role of muscle-secreted factors. Obesity (Silver Spring) 2025; 33:435-451. [PMID: 39948829 PMCID: PMC11897867 DOI: 10.1002/oby.24223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 03/14/2025]
Abstract
As the prevalence of obesity and metabolic disease continues to climb, the need for effective therapeutic interventions remains high. The growth of skeletal muscle (SkM) greatly influences systemic metabolism across the whole body, making this tissue an important therapeutic target to combat the rise of metabolic dysfunction. Transgenic rodent models of targeted SkM growth exhibit profound improvements in various remote tissues, including adipose tissue and the liver. It is currently unclear how selective stimulation of SkM growth alters the metabolism of distant tissues; however, evidence suggests that muscle-secreted factors may be involved. Here, we aim to provide basic biomedical researchers with a summary of the current knowledge regarding various muscle-secreted factors regulated by anabolic pathways and proteins in SkM, as well as their systemic metabolic effects, to implicate them in the whole-body metabolic effects of SkM growth. In this review, we also identify several knowledge gaps in this field, future directions of investigation, and implications for therapeutic interventions such as resistance exercise and pharmacology.
Collapse
Affiliation(s)
- Mitchell J. Sammut
- School of Kinesiology, Faculty of Health SciencesWestern UniversityLondonOntarioCanada
| | - Benjamin R. Thorne
- School of Kinesiology, Faculty of Health SciencesWestern UniversityLondonOntarioCanada
| | - C. W. James Melling
- School of Kinesiology, Faculty of Health SciencesWestern UniversityLondonOntarioCanada
- Department of Physiology & Pharmacology, Schulich School of Medicine & DentistryWestern UniversityLondonOntarioCanada
| |
Collapse
|
6
|
Smith WB, Nguyen D, Clough T, Schofield J, Kagan MR, Kompa J, He Y, Maratos-Flier E, Jamontt J, Vong L, Schwartzkopf CD, Layne JD, Usera AR, O'Donnell CJ, Heldwein KA, Streeper RS, Goldfine AB. A Growth Differentiation Factor 15 Receptor Agonist in Randomized Placebo-Controlled Trials in Healthy or Obese Persons. J Clin Endocrinol Metab 2025; 110:771-786. [PMID: 39148430 DOI: 10.1210/clinem/dgae550] [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: 05/31/2024] [Revised: 07/20/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Growth differentiation factor 15 (GDF15), a divergent member of the TGF-β superfamily, signals via the hindbrain glial-derived neurotrophic factor receptor alpha-like and rearranged during transfection receptor co-receptor (GFRAL-RET) complex. In nonclinical species, GDF15 is a potent anorexigen leading to substantial weight loss. MBL949 is a half-life extended recombinant human GDF15 dimer. METHODS MBL949 was evaluated in multiple nonclinical species, and then in humans, in 2 randomized and placebo-controlled clinical trials. In the phase 1, first-in-human, single ascending dose trial, MBL949 or placebo was injected subcutaneously to overweight and obese healthy volunteers (n = 65) at doses ranging from 0.03 to 20 mg. In phase 2, MBL949 or placebo was administered subcutaneously every other week for a total of 8 doses to obese participants (n = 126) in 5 different dose regimens predicted to be efficacious based on data from the phase 1 trial. RESULTS In nonclinical species, MBL949 was generally safe and effective with reduced food intake and body weight in mice, rats, dogs, and monkeys. Weight loss was primarily from reduced fat, and metabolic endpoints improved. A single ascending dose study in overweight or obese healthy adults demonstrated mean terminal half-life of 18 to 22 days and evidence of weight loss at the higher doses. In the phase 2, weight loss was minimal following biweekly dosing of MBL949 for 14 weeks. MBL949 was safe and generally tolerated in humans over the dose range tested, adverse events of the gastrointestinal system were the most frequent observed. CONCLUSION The prolonged half-life of MBL949 supports biweekly dosing in patients. MBL949 had an acceptable safety profile. The robust weight loss observed in nonclinical species did not translate to weight loss efficacy in humans.
Collapse
Affiliation(s)
- William B Smith
- Alliance for Multispecialty Research, LLC, Knoxville, TN 37909, USA
| | - David Nguyen
- Altasciences Clinical Los Angeles, Inc., Cypress, CA 90630, USA
| | - Timothy Clough
- Novartis Biomedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Jül Schofield
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | - Mark R Kagan
- Novartis Pharmaceuticals Corporation, Cardiovascular, Renal and Metabolic, East Hanover, NJ 07936, USA
| | - Jill Kompa
- Novartis Pharmaceuticals Corporation, Cardiovascular, Renal and Metabolic, East Hanover, NJ 07936, USA
| | - YanLing He
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | | | - Joanna Jamontt
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | - Linh Vong
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | - Chad D Schwartzkopf
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | - Joseph D Layne
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | - Aimee R Usera
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | | | - Kurt A Heldwein
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | - Ryan S Streeper
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| | - Allison B Goldfine
- Novartis Biomedical Research, Cardiovascular, Renal and Metabolic, Cambridge, MA 02139, USA
| |
Collapse
|
7
|
Flaherty S, Song L, Albuquerque B, Rinaldi A, Piper M, Shanthappa D, Chen X, Stansfield J, Asano S, Pashos E, Ross T, Jagarlapudi S, Sheikh A, Zhang B, Wu Z. GDF15 Neutralization Ameliorates Muscle Atrophy and Exercise Intolerance in a Mouse Model of Mitochondrial Myopathy. J Cachexia Sarcopenia Muscle 2025; 16:e13715. [PMID: 39976232 PMCID: PMC11840706 DOI: 10.1002/jcsm.13715] [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: 12/21/2023] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Primary mitochondrial myopathies (PMMs) are disorders caused by mutations in genes encoding mitochondrial proteins and proteins involved in mitochondrial function. PMMs are characterized by loss of muscle mass and strength as well as impaired exercise capacity. Growth/Differentiation Factor 15 (GDF15) was reported to be highly elevated in PMMs and cancer cachexia. Previous studies have shown that GDF15 neutralization is effective in improving skeletal muscle mass and function in cancer cachexia. It remains to be determined if the inhibition of GDF15 could be beneficial for PMMs. The purpose of the present study is to assess whether treatment with a GDF15 neutralizing antibody can alleviate muscle atrophy and physical performance impairment in a mouse model of PMM. METHODS The effects of GDF15 neutralization on PMM were assessed using PolgD257A/D257A (POLG) mice. These mice express a proofreading-deficient version of the mitochondrial DNA polymerase gamma, leading to an increased rate of mutations in mitochondrial DNA (mtDNA). These animals display increased circulating GDF15 levels, reduced muscle mass and function, exercise intolerance, and premature aging. Starting at 9 months of age, the mice were treated with an anti-GDF15 antibody (mAB2) once per week for 12 weeks. Body weight, food intake, body composition, and muscle mass were assessed. Muscle function and exercise capacity were evaluated using in vivo concentric max force stimulation assays, forced treadmill running and voluntary home-cage wheel running. Mechanistic investigations were performed via muscle histology, bulk transcriptomic analysis, RT-qPCR and western blotting. RESULTS Anti-GDF15 antibody treatment ameliorated the metabolic phenotypes of the POLG animals, improving body weight (+13% ± 8%, p < 0.0001), lean mass (+13% ± 15%, p < 0.001) and muscle mass (+35% ± 24%, p < 0.001). Additionally, the treatment improved skeletal muscle max force production (+35% ± 43%, p < 0.001) and exercise performance, including treadmill (+40% ± 29%, p < 0.05) and voluntary wheel running (+320% ± 19%, p < 0.05). Mechanistically, the beneficial effects of GDF15 neutralization are linked to the reversal of the transcriptional dysregulation in genes involved in autophagy and proteasome signalling. The treatment also appears to dampen glucocorticoid signalling by suppressing circulating corticosterone levels in the POLG animals. CONCLUSIONS Our findings highlight the potential of GDF15 neutralization with a monoclonal antibody as a therapeutic avenue to enhance physical performance and mitigate adverse clinical outcomes in patients with PMM.
Collapse
Affiliation(s)
- Stephen E. Flaherty
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Obesity and ComplicationsEli LillyBostonMassachusettsUSA
| | - LouJin Song
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bina Albuquerque
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Anthony Rinaldi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Program Mamager, Preclinical Sciences, ToxicologyVertex PharmaceuticalsBostonMassachusettsUSA
| | - Mary Piper
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | | | - Xian Chen
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - John Stansfield
- Biostatistics, Early Clinical DevelopmentPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Shoh Asano
- Inflammation and Immunology Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Evanthia Pashos
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Trenton Thomas Ross
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Srinath Jagarlapudi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Abdul Sheikh
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bei Zhang
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Zhidan Wu
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| |
Collapse
|
8
|
Dantzer R, Chelette B, Vichaya EG, West AP, Grossberg A. The metabolic basis of cancer-related fatigue. Neurosci Biobehav Rev 2025; 169:106035. [PMID: 39892436 PMCID: PMC11866516 DOI: 10.1016/j.neubiorev.2025.106035] [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: 10/22/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Although we are all familiar with the sensation of fatigue, there are still profound divergences on what it represents and its mechanisms. Fatigue can take various forms depending on the condition in which it develops. Cancer-related fatigue is considered a symptom of exhaustion that is often present at the time of diagnosis, increases in intensity during cancer therapy, and does not always recede after completion of treatment. It is usually attributed to the inflammation induced by damage-associated molecular patterns released by tumor cells during cancer progression and in response to its treatment. In this review, we argue that it is necessary to go beyond the symptoms of fatigue to understand its nature and mechanisms. We propose to consider fatigue as a psychobiological process that regulates the behavioral activities an organism engages in to satisfy its needs, according to its physical ability to do so and to the capacity of its intermediary metabolism to exploit the resources procured by these activities. This last aspect is critical as it implies that these metabolic aspects need to be considered to understand fatigue. Based on the findings we have accumulated over several years of studying fatigue in diverse murine models of cancer, we show that energy metabolism plays a key role in the development and persistence of this condition. Cancer-related fatigue is dependent on the energy requirements of the tumor and the negative impact of cancer therapy on the mitochondrial function of the host. When inflammation is present, it adds to the organism's energy expenses. The organism needs to adjust its metabolism to the different forms of cellular stress it experiences thanks to specialized communication factors known as mitokines that act locally and at a distance from the cells in which they are produced. They induce the subjective, behavioral, and metabolic components of fatigue by acting in the brain. Therefore, the targeting of mitokines and their brain receptors offers a window of opportunity to treat fatigue when it is no longer adaptive but an obstacle to the quality of life of cancer survivors.
Collapse
Affiliation(s)
- Robert Dantzer
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Brandon Chelette
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elisabeth G Vichaya
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, USA
| | | | - Aaron Grossberg
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
9
|
James E, Frampton J, Murphy KG, Chambers ES. Effects of Acute Exercise and Carbohydrate Intake on Plasma GDF-15 Levels and Its Association With Appetite Regulation. J Endocr Soc 2025; 9:bvaf013. [PMID: 39902404 PMCID: PMC11788510 DOI: 10.1210/jendso/bvaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Indexed: 02/05/2025] Open
Abstract
Background Growth differentiation factor 15 (GDF-15) is a potential therapeutic target for obesity due to its role in appetite suppression. Although acute exercise stimulates GDF-15 secretion, its relationship with appetite regulation remains unclear. It is also unknown whether preexercise carbohydrate intake would affect GDF-15 responses. This study aimed to examine the effects of acute exercise and carbohydrate intake on GDF-15 secretion and its potential links to appetite regulation. Methods In a secondary analysis of a randomized crossover trial, 12 healthy males completed four 120-minute trial conditions: (1) control (water) with rest, (2) control with exercise (0-30 minutes at ∼75% of maximal oxygen uptake), (3) carbohydrate (75 g maltodextrin) with rest, and (4) carbohydrate with exercise. Plasma GDF-15 levels were measured at 0, 30, 60, and 120 minutes, alongside subjective appetite ratings using visual analog scales. Energy intake was measured at the end of each trial condition with an ad libitum meal. Results Time-averaged area under the curve analysis showed that neither exercise [34 pg/mL (95% confidence interval [CI], -2-69 pg/mL); P = .062) nor carbohydrate intake [10 pg/mL (95% CI, -39-58 pg/mL); P = .673] independently or interactively (P = .283) affected GDF-15 levels. Exercise induced a delayed independent increase in GDF-15 at 120 minutes [55 pg/mL (95% CI, 18-94 pg/mL); P = .008]. No significant associations were found between GDF-15 levels and subjective appetite ratings or energy intake. Conclusion A 30-minute bout of high-intensity exercise induces a delayed increase in GDF-15 levels, which is not affected by carbohydrate intake. Physiological GDF-15 responses to acute exercise display no association with markers of appetite regulation.
Collapse
Affiliation(s)
- Ellen James
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - James Frampton
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Kevin G Murphy
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Edward S Chambers
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| |
Collapse
|
10
|
Hes C, Gui LT, Bay A, Alvarez F, Katz P, Paul T, Bozadjieva-Kramer N, Seeley RJ, Piccirillo CA, Sabatini PV. GDNF family receptor alpha-like (GFRAL) expression is restricted to the caudal brainstem. Mol Metab 2025; 91:102070. [PMID: 39608751 PMCID: PMC11650321 DOI: 10.1016/j.molmet.2024.102070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVE Growth differentiation factor 15 (GDF15) acts on the receptor dimer of GDNF family receptor alpha-like (GFRAL) and Rearranged during transfection (RET). While Gfral-expressing cells are known to be present in the area postrema and nucleus of the solitary tract (AP/NTS) located in the brainstem, the presence of Gfral-expressing cells in other sites within the central nervous system and peripheral tissues is not been fully addressed. Our objective was to thoroughly investigate whether GFRAL is expressed in peripheral tissues and in brain sites different from the brainstem. METHODS From Gfral:eGFP mice we collected tissue from 12 different tissues, including brain, and used single molecule in-situ hybridizations to identify cells within those tissues expressing Gfral. We then contrasted the results with human Gfral-expression by analyzing publicly available single-cell RNA sequencing data. RESULTS In mice we found readably detectable Gfral mRNA within the AP/NTS but not within other brain sites. Within peripheral tissues, we failed to detect any Gfral-labelled cells in the vast majority of examined tissues and when present, were extremely rare. Single cell sequencing of human tissues confirmed GFRAL-expressing cells are detectable in some sites outside the AP/NTS in an extremely sparse manner. Importantly, across the utilized methodologies, smFISH, genetic Gfral reporter mice and scRNA-Seq, we failed to detect Gfral-labelled cells with all three. CONCLUSIONS Through highly sensitive and selective technologies we show Gfral expression is overwhelmingly restricted to the brainstem and expect that GDF15 and GFRAL-based therapies in development for cancer cachexia will specifically target AP/NTS cells.
Collapse
Affiliation(s)
- Cecilia Hes
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada; Division of Experimental Medicine, Department of Medicine, McGill University, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada
| | - Lu Ting Gui
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada; Integrated Program in Neuroscience, Department of Medicine, McGill University, Room 302 Irving Ludmer Building, 1033 Pine Ave. W. Montreal, QC, H3A 1A1, Canada
| | - Alexandre Bay
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada
| | - Fernando Alvarez
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada
| | - Pierce Katz
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada; Integrated Program in Neuroscience, Department of Medicine, McGill University, Room 302 Irving Ludmer Building, 1033 Pine Ave. W. Montreal, QC, H3A 1A1, Canada
| | - Tanushree Paul
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA; Veterans Affairs Ann Arbor Healthcare System, Research Service, 2215 Fuller Rd, Ann Arbor, MI, 48105, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA
| | - Ciriaco A Piccirillo
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada; Department of Microbiology and Immunology, Department of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A 2B4, Canada; Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada; Program in Infectious Diseases and Immunology in Global Health, Research Institute of the McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada
| | - Paul V Sabatini
- Research Institute of the McGill University Health Centre, McGill University Health Centre, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada; Division of Experimental Medicine, Department of Medicine, McGill University, 1001 boulevard de Decarie, Montreal, QC, H4A 3J1, Canada; Integrated Program in Neuroscience, Department of Medicine, McGill University, Room 302 Irving Ludmer Building, 1033 Pine Ave. W. Montreal, QC, H3A 1A1, Canada.
| |
Collapse
|
11
|
Wang X, Zhang G. The mitochondrial integrated stress response: A novel approach to anti-aging and pro-longevity. Ageing Res Rev 2025; 103:102603. [PMID: 39608727 DOI: 10.1016/j.arr.2024.102603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The ISR is a cellular signaling pathway that responds to various physiological changes and types of stimulation. The mitochondrial integrated stress response (ISRmt) is a stress response specific to mitochondria which is initiated by eIF2α phosphorylation and is responsive to mitochondrial stressors. The ISRmt triggers diverse metabolic responses reliant on activating transcription factor 4 (ATF4). The preliminary phases of ISRmt can provoke an adaptive stress response that antagonizes age-related diseases and promotes longevity. In this review, we provide an overview of the molecular mechanisms of the ISRmt, with a particular focus on its potential as a therapeutic target for age-related disease and the promotion of longevity.
Collapse
Affiliation(s)
- Xiaoding Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China.
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China.
| |
Collapse
|
12
|
Rao Z, Geng X, Huang P, Wei Q, Liu S, Qu C, Zhao J. Housing temperature influences exercise-induced glucose regulation and expression of exerkines in mice. Exp Physiol 2024. [PMID: 39721028 DOI: 10.1113/ep092319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
The impact of housing temperature on exercise-induced metabolic adaptations is not well understood, despite extensive research on the benefits of exercise for metabolic health. The aim of this study was to elucidate how housing temperatures influence the molecular responses and metabolic benefits of exercise in mice. Male C57BL/6N mice were housed at either room temperature (RT, 21°C) or in a thermoneutral environment (TN, 29°C) and subjected to either a 6-week or acute exercise regimen. The results demonstrated that chronic exercise in TN conditions significantly improved glucose tolerance, whereas no such improvement was observed in RT conditions. Exercise reduced adipocyte size in inguinal and epididymal white adipose tissue in RT conditions, but no significant exercise-induced browning of inguinal white adipose tissue was detected at either housing temperature. Additionally, housing temperature predominantly influenced key metabolic proteins in skeletal muscle, with exercise and temperature exhibiting interactive effects on glycogen synthase, Glut4 and Pgc-1α. Moreover, the regulation of exerkines, including Fgf21, fetuin-A, irisin, Gdf15, spexin and apelin, was temperature dependent after both long-term and acute exercise. Notably, expression of Metrnl was consistently upregulated in skeletal muscle after long-term exercise in both RT and TN environments, but was downregulated after acute exercise. These findings highlight that environmental temperature critically modulates the metabolic benefits of exercise and the expression of exerkines. The results of this study suggest that conventional RT conditions might obscure the full metabolic effects of exercise. We recommend the use of TN conditions in future research to reduce confounding factors and provide a more accurate assessment of the metabolic benefits of exercise.
Collapse
Affiliation(s)
- Zhijian Rao
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
- College of Physical Education, Shanghai Normal University, Shanghai, China
| | - Xue Geng
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
| | - Peng Huang
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
| | - Qiangman Wei
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
| | - Shijie Liu
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
| | - Chaoyi Qu
- College of Physical Education, Hebei Normal University, Hebei, China
| | - Jiexiu Zhao
- Exercise Biological Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
13
|
Isik FI, Thomson S, Cueto JF, Spathos J, Breit SN, Tsai VWW, Brown DA, Finney CA. A systematic review of the neuroprotective role and biomarker potential of GDF15 in neurodegeneration. Front Immunol 2024; 15:1514518. [PMID: 39737171 PMCID: PMC11682991 DOI: 10.3389/fimmu.2024.1514518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Neurodegeneration is characteristically multifaceted, with limited therapeutic options. One of the chief pathophysiological mechanisms driving these conditions is neuroinflammation, prompting increasing clinical interest in immunomodulatory agents. Growth differentiation factor 15 (GDF15; previously also called macrophage inhibitory cytokine-1 or MIC-1), an anti-inflammatory cytokine with established neurotrophic properties, has emerged as a promising therapeutic agent in recent decades. However, methodological challenges and the delayed identification of its specific receptor GFRAL have hindered research progress. This review systematically examines literature about GDF15 in neurodegenerative diseases and neurotrauma. The evidence collated in this review indicates that GDF15 expression is upregulated in response to neurodegenerative pathophysiology and increasing its levels in preclinical models typically improves outcomes. Key knowledge gaps are addressed for future investigations to foster a more comprehensive understanding of the neuroprotective effects elicited by GDF15.
Collapse
Affiliation(s)
- Finula I. Isik
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Shannon Thomson
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - John F. Cueto
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Jessica Spathos
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Samuel N. Breit
- St. Vincent’s Centre for Applied Medical Research, St. Vincent’s Hospital and Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Vicky W. W. Tsai
- St. Vincent’s Centre for Applied Medical Research, St. Vincent’s Hospital and Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - David A. Brown
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Western Sydney Local Health District, Institute for Clinical Pathology and Medical Research, NSW Health Pathology, Sydney, NSW, Australia
| | - Caitlin A. Finney
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
14
|
Liu C, Wong PY, Wang Q, Wong HY, Huang T, Cui C, Zhang N, Cheung WH, Wong RMY. Short-chain fatty acids enhance muscle mass and function through the activation of mTOR signalling pathways in sarcopenic mice. J Cachexia Sarcopenia Muscle 2024; 15:2387-2401. [PMID: 39482890 PMCID: PMC11634463 DOI: 10.1002/jcsm.13573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Sarcopenia is a prevalent muscle disorder in old people leading to higher fracture rate, mortality, and other adverse clinical outcomes. Evidence indicates that short-chain fatty acids (SCFAs), which are beneficial gut microbial metabolites, were reduced in old people with sarcopenia. This study aimed to determine whether the use of SCFAs as a supplement can be a therapeutic strategy of sarcopenia in a pre-clinical model. METHODS Seven-month-old pre-sarcopenic senescent accelerated mouse prone 8 (SAMP8) mice received daily SCFAs cocktail (acetate, butyrate, and propionate) for 3 months. Age-matched senescence accelerated mouse resistant 1 (SAMR1) and SAMP8 mice receiving sodium-matched drinking water were control groups. The gut microbiota composition analysis of aged mice with or without sarcopenia was conducted by 16S rDNA sequencing. Gut barrier-related proteins and lipopolysaccharide (LPS) concentration were biomarkers of gut permeability. Colon inflammation levels, circulatory SCFAs concentration, muscle quality, function, and underlying pathways were detected by cell number counting, RT-qPCR, gas chromatography-mass spectrometry, measurements of muscle wet weight and grip strength, ex vivo functional test, treadmill endurance test, transcriptomic sequencing, morphological and immunofluorescent staining, as well as western blot. To investigate the role of mTOR signalling pathways in SCFAs treatment, C2C12 myotubes were treated with rapamycin. RESULTS Aged SAMP8 mice had different microbiota composition, and lower serum butyric acid compared with SAMR1 mice (P < 0.05). SCFAs treatment reversed the increment of colon inflammation (2.8-fold lower of il-1β) and gut barrier permeability (1.7-fold lower of LPS) in SAMP8 mice. Increased muscle mass, myofibre cross-sectional area, grip strength, twitch and tetanic force were found in SCFAs-treated mice compared with control SAMP8 mice (P < 0.05). Anti-fatigue capacity (1.6-fold) and muscle glycogen (2-fold) also improved after SCFAs treatment (P < 0.05). Transcriptomic analysis showed that AMPK, insulin, and mTOR pathways were involved in SCFAs treatment (P < 0.05). Regulation of AKT/mTOR/S6K1 and AMPK/PGC1α pathways were found. SCFAs attenuated fat infiltration and improved mitochondria biogenesis of atrophic muscle. In vitro studies indicated that SCFAs inhibited FoxO3a/Atrogin1 and activated mTOR pathways to improve myotube growth (P < 0.05), and rapamycin attenuated the effect of SCFAs through the inhibition of mTOR pathways. CONCLUSIONS This study demonstrated that bacterial metabolites SCFAs could attenuate age-related muscle loss and dysfunction, and protein synthesis-related mTOR signalling pathways were involved both in vivo and in vitro.
Collapse
Affiliation(s)
- Chaoran Liu
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Pui Yan Wong
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Qianjin Wang
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Hei Yuet Wong
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Tao Huang
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Can Cui
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Ning Zhang
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Wing Hoi Cheung
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Ronald Man Yeung Wong
- Department of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
15
|
Arbeau M, Baranowski BJ, Jeromson S, Bellucci A, Akcan M, Trang S, Eisner K, Medak KD, Wright DC. GDF15 associates with, but is not responsible for, exercise-induced increases in corticosterone and indices of lipid utilization in mice. J Appl Physiol (1985) 2024; 137:1512-1523. [PMID: 39480267 PMCID: PMC11687845 DOI: 10.1152/japplphysiol.00519.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Growth differentiation factor 15 (GDF15) is a stress-induced cytokine that increases with exercise and is thought to increase corticosterone and lipid utilization. How postexercise nutrient availability impacts GDF15 and the physiological role that GDF15 plays during and/or in the recovery from exercise has not been elucidated. The purpose of this investigation was to examine how postexercise nutrient availability impacts GDF15 and to use this as a model to explore associations between GDF15, corticosterone, and indices of lipid and carbohydrate metabolism. In addition, we explored the causality of these relationships using GDF15-deficient mice. Male and female C57BL/6J mice ran for 2 hours on a treadmill and were euthanized immediately or 3 hours after exercise with or without access to a chow diet. In both sexes, circulating concentrations of GDF15, corticosterone, nonesterified fatty acids (NEFA), and beta-hydroxybutyrate (BHB) were higher immediately postexercise and remained elevated when food was withheld during the recovery period. While serum GDF15 was positively associated with corticosterone, BHB, and NEFA, increases in these factors were similar in wild-type and GDF15-/- mice following exercise. The lack of a genotype effect was not explained by differences in insulin, glucagon, or epinephrine after exercise. Our findings provide evidence that while GDF15 is associated with increases in corticosterone and indices of lipid utilization this is not a causal relationship.NEW & NOTEWORTHY Circulating growth differentiation factor 15 (GDF15) increases during exercise, but the physiological role that it plays has not been elucidated. Recent data suggest that GDF15 regulates corticosterone and lipid utilization. Here we demonstrate that postexercise nutrient availability influences GDF15 in the recovery from exercise and GDF15 is associated with corticosterone and indices of lipid utilization. However, the associations were not causal as exercise-induced increases in fatty acids, beta-hydroxybutyrate, and corticosterone were intact in GDF15-/- mice.
Collapse
Affiliation(s)
- Meagan Arbeau
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Bradley J Baranowski
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Stewart Jeromson
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Annalaura Bellucci
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Michael Akcan
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Serena Trang
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Katelyn Eisner
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Kyle D Medak
- Deparment of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David C Wright
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Jeromson S, Akcan M, Baranowski B, Arbeau M, Bellucci A, Wright DC. Daily GDF15 treatment has sex-specific effects on body weight and food intake and does not enhance the effects of voluntary physical activity in mice. J Physiol 2024; 602:6813-6826. [PMID: 39521949 DOI: 10.1113/jp287256] [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: 07/14/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Growth differentiation factor 15 (GDF15) is a stress-induced cytokine that suppresses food intake and causes weight loss. GDF15 also reduces voluntary physical activity and, thus, it is not clear whether combining GDF15 with exercise will be beneficial or if reductions in food intake would be offset by decreases in physical activity. We investigated how GDF15 treatment combined with voluntary wheel running (VWR) would impact weight gain, food intake, adiposity and indices of metabolic health in mice. High-fat fed male and female mice underwent daily GDF15 treatments and were given access to voluntary running wheels, or not, for 11 days. In both sexes, VWR prevented weight gain. In males, GDF15 reduced food intake, as well as attenuated weight gain and the accumulation of adipose tissue, with no additional effect of VWR. In female mice, GDF15 did not impact body weight gain or body composition. GDF15 acutely reduced food intake in female mice but this was followed by a period of rebound hyperphagia and consequently GDF15 did not reduce total food intake in female mice. GDF15 treatment reduced wheel running distance in both sexes. There were main effects of VWR to improve glucose tolerance in female but not male mice. These findings show that GDF15 has sex-specific effects on food intake and consequently weight gain and adiposity. There is no added benefit of combining GDF15 and voluntary physical activity for weight loss. Adaptive responses to acute caloric restriction induced by GDF15 might limit its effectiveness as a weight loss tool in females. KEY POINTS: GDF15 is a stress-induced signalling factor that reduces food intake and voluntary physical activity. It is not known whether combining GDF15 treatment with voluntary wheel running would impart beneficial combined effects in attenuating weight gain and the accumulation of adipose tissue. In the present study, we demonstrate that GDF15 reduces food intake and prevents weight gain in male but not female mice consuming a high-fat diet and also that combining GDF15 with voluntary wheel running (VWR) does not lead to a greater dampening of weight gain. In female mice, GDF15 acutely reduced food intake, but this was followed by a period of rebound hyperphagia resulting in no differences in total food intake. In both sexes, VWR was equivalent, or superior to GDF15 in preventing weight gain.
Collapse
Affiliation(s)
- Stewart Jeromson
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Michael Akcan
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Bradley Baranowski
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Meagan Arbeau
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Annalaura Bellucci
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - David C Wright
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
17
|
Huang Q, Monzel AS, Rausser S, Haahr R, Devine J, Liu CC, Kelly C, Thompson E, Kurade M, Michelson J, Li S, Engelstad K, Tanji K, Lauriola V, Wang T, Wang S, Marsland AL, Kaufman BA, St-Onge MP, Sloan R, Juster RP, Gouspillou G, Hirano M, Picard M, Trumpff C. The Energetic Stress Marker GDF15 is Induced by Acute Psychosocial Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590241. [PMID: 38659958 PMCID: PMC11042343 DOI: 10.1101/2024.04.19.590241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
GDF15 (growth differentiation factor 15) is a marker of cellular and mitochondrial energetic stress linked to physical-mental illness, aging, and mortality. Here, we describe the psychobiological regulation of plasma and saliva GDF15 in four human studies including 3,599 samples from 148 healthy individuals. We report two main observations establishing GDF15 as a novel tractable biomarker of psychosocial stress. 1) In two experimental laboratory studies, socio-evaluative stress rapidly elevates GDF15 and lactate, two molecular markers of energetic/reductive stress. 2) Similar to other stress-related metabolic hormones, we also find that saliva GDF15 exhibit a robust awakening response, being highest at the time of waking up and declining by ~42-92% within 30-45 minutes. These data position GDF15 as a dynamic biomarker of psychosocial stress accessible in human blood and saliva, pointing towards a shared psychobiological pathway linking mental and mitochondrial energetic stress. These foundational observations open the door to large-scale studies using GDF15 to non-invasively probe how acute psychosocial factors promote cellular and mitochondrial and energetic stress contributing to the stress-disease cascade across the lifespan.
Collapse
Affiliation(s)
- Qiuhan Huang
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S. Monzel
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Shannon Rausser
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Rachel Haahr
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Devine
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Cynthia C. Liu
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Catherine Kelly
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth Thompson
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Mangesh Kurade
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeremy Michelson
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Shufang Li
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Kris Engelstad
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Kurenai Tanji
- Department of pathology and cell biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Vincenzo Lauriola
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Tian Wang
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Shuang Wang
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Anna L Marsland
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brett A Kaufman
- Department of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA United States
| | - Marie-Pierre St-Onge
- Division of General Medicine and Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, USA
| | - Richard Sloan
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Robert-Paul Juster
- Department of Psychiatry and Addiction, University of Montreal, Montreal, QC, Canada
| | - Gilles Gouspillou
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC, Canada
| | - Michio Hirano
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
18
|
Mulcahy MC, El Habbal N, Redd JR, Sun H, Gregg BE, Bridges D. GDF15 Knockout Does Not Substantially Impact Perinatal Body Weight or Neonatal Outcomes in Mice. Endocrinology 2024; 165:bqae143. [PMID: 39445824 PMCID: PMC11577612 DOI: 10.1210/endocr/bqae143] [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: 05/14/2024] [Revised: 08/08/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024]
Abstract
Growth differentiation factor-15 (GDF15) increases in circulation during pregnancy and has been implicated in food intake, weight loss, complications of pregnancy, and metabolic illness. We used a Gdf15 knockout mouse model (Gdf15-/-) to assess the role of GDF15 in body weight regulation and food intake during pregnancy. We found that Gdf15-/- dams consumed a similar amount of food and gained comparable weight during the course of pregnancy compared with Gdf15+/+ dams. Insulin sensitivity on gestational day 16.5 was also similar between genotypes. In the postnatal period, litter size and survival rates were similar between genotypes. There was a modest reduction in birth weight of Gdf15-/- pups, but this difference was no longer evident from postnatal day 3.5 to 14.5. We observed no detectable differences in milk volume production or milk fat percentage. These data suggest that GDF15 is dispensable for changes in food intake, and body weight as well as insulin sensitivity during pregnancy in a mouse model.
Collapse
Affiliation(s)
- Molly C Mulcahy
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Noura El Habbal
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- School of Health Professions, New York Institute of Technology, Old Westbury, New York, NY 11568, USA
| | - JeAnna R Redd
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Haijing Sun
- Department of Pediatric Endocrinology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Brigid E Gregg
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Pediatric Endocrinology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| |
Collapse
|
19
|
Okada TE, Jeromson S, Rathwell S, Wright DC, Bomhof MR. Aerobic exercise elevates perceived appetite but does not modify energy intake over a 3-day postexercise period: A pilot study. Physiol Rep 2024; 12:e70066. [PMID: 39328151 PMCID: PMC11427932 DOI: 10.14814/phy2.70066] [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: 07/06/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
While a low degree of energy compensation is typically reported over the 24 h following a session of exercise, the prolonged impact of a bout of exercise on energy intake remains unclear. To overcome the challenge associated with accurately measuring energy intake in a free-living environment, this study employed the use of a meal replacement beverage to assess the 3 day impact of an exercise session on energy intake. In a randomized, crossover study, 14 participants (8 male, 6 female) completed two trials: (1) EX: 75 min exercise on a motorized treadmill (75% VO2peak); and (2) SED: 75 min sedentary control session. Each condition was followed by 3 days of exclusive ad libitum consumption of a meal replacement beverage. Appetite-regulating hormones, subjective appetite, energy intake, and energy expenditure were assessed. Exercise transiently suppressed the orexigenic hormone acyl-ghrelin (p < 0.05) and elevated the appetite-supressing hepatokine GDF-15 (p < 0.05). Despite these acute changes, overall perceived appetite was elevated over the 3 day assessment period with exercise (p < 0.05). No increase in energy intake or change in postexercise physical activity patterns were observed. One acute session of moderate to vigorous exercise is unlikely to affect short-term, three-day energy balance in healthy individuals.
Collapse
Affiliation(s)
- Tetsuro E. Okada
- Department of Kinesiology and Physical EducationUniversity of LethbridgeLethbridgeAlbertaCanada
| | - Stewart Jeromson
- School of KinesiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Scott Rathwell
- Department of Kinesiology and Physical EducationUniversity of LethbridgeLethbridgeAlbertaCanada
| | - David C. Wright
- School of KinesiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Faculty of Land and Food SystemsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| | - Marc R. Bomhof
- Department of Kinesiology and Physical EducationUniversity of LethbridgeLethbridgeAlbertaCanada
| |
Collapse
|
20
|
Labour A, Lac M, Frassin L, Lair B, Murphy E, Maslo C, Monbrun L, Calmy ML, Marquès M, Viguerie N, Tavernier G, Gourdy P, O'Gorman D, Montastier E, Laurens C, Montagner A, Moro C. GDF15 is dispensable for the insulin-sensitizing effects of chronic exercise. Cell Rep 2024; 43:114577. [PMID: 39096490 DOI: 10.1016/j.celrep.2024.114577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/05/2024] Open
Abstract
Growth and differentiation factor 15 (GDF15) has recently emerged as a weight loss and insulin-sensitizing factor. Growing evidence also supports a role for GDF15 as a physiological, exercise-induced stress signal. Here, we tested whether GDF15 is required for the insulin-sensitizing effects of exercise in mice and humans. At baseline, both under a standard nutritional state and high-fat feeding, GDF15 knockout (KO) mice display normal glucose tolerance, systemic insulin sensitivity, maximal speed, and endurance running capacity when compared to wild-type littermates independent of sex. When submitted to a 4-week exercise training program, both lean and obese wild-type and GDF15 KO mice similarly improve their endurance running capacity, glucose tolerance, systemic insulin sensitivity, and peripheral glucose uptake. Insulin-sensitizing effects of exercise training were also unrelated to changes in plasma GDF15 in humans. In summary, we here show that GDF15 is dispensable for the insulin-sensitizing effects of chronic exercise.
Collapse
Affiliation(s)
- Axel Labour
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Marlène Lac
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Lucas Frassin
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Benjamin Lair
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Enda Murphy
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Claire Maslo
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Laurent Monbrun
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Marie-Lou Calmy
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Marie Marquès
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Nathalie Viguerie
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Geneviève Tavernier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Pierre Gourdy
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France; Department of Diabetology, Toulouse University Hospital, Toulouse, France
| | - Donal O'Gorman
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Emilie Montastier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France; Department of Endocrinology, Metabolic Diseases and Nutrition, Toulouse University Hospital, Toulouse, France
| | - Claire Laurens
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Alexandra Montagner
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France
| | - Cedric Moro
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Toulouse, France.
| |
Collapse
|
21
|
Qiao R, Guo J, Zhang C, Wang S, Fang J, Geng R, Kang SG, Huang K, Tong T. Diabetes-induced muscle wasting: molecular mechanisms and promising therapeutic targets. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39049742 DOI: 10.1080/10408398.2024.2382348] [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: 07/27/2024]
Abstract
Diabetes has become a serious public health crisis, presenting significant challenges to individuals worldwide. As the largest organ in the human body, skeletal muscle is a significant target of this chronic disease, yet muscle wasting as a complication of diabetes is still not fully understood and effective treatment methods have yet to be developed. Here, we discuss the targets involved in inducing muscle wasting under diabetic conditions, both validated targets and emerging targets. Diabetes-induced skeletal muscle wasting is known to involve changes in various signaling molecules and pathways, such as protein degradation pathways, protein synthesis pathways, mitochondrial function, and oxidative stress inflammation. Recent studies have shown that some of these present potential as promising therapeutic targets, including the neuregulin 1/epidermal growth factor receptor family, advanced glycation end-products, irisin, ferroptosis, growth differentiation factor 15 and more. This study's investigation and discussion of such pathways and their potential applications provides a theoretical basis for the development of clinical treatments for diabetes-induced muscle wasting and a foundation for continued focus on this disease.
Collapse
Affiliation(s)
- Ruixue Qiao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Chengmei Zhang
- Guizhou Academy of Testing and Analysis, Guiyang, The People's Republic of China
| | - Sirui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Jingjing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Ruixuan Geng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, The People's Republic of China
- Beijing Laboratory for Food Quality and Safety, Beijing, The People's Republic of China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, The People's Republic of China
- Beijing Laboratory for Food Quality and Safety, Beijing, The People's Republic of China
| |
Collapse
|
22
|
Fang S, Ji Y, Shen Y, Yang S, Zhang H, Xin W, Shi W, Chen W. TET3 Contributes to Exercise-Induced Functional Axon Regeneration and Visual Restoration. Adv Biol (Weinh) 2024:e2400145. [PMID: 39007414 DOI: 10.1002/adbi.202400145] [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: 03/13/2024] [Revised: 05/27/2024] [Indexed: 07/16/2024]
Abstract
Axons have intrinsically poor regenerative capacity in the mature central nervous system (CNS), leading to permanent neurological impairments in individuals. There is growing evidence that exercise is a powerful physiological intervention that can obviously enhance cell rejuvenate capacity, but its molecular mechanisms that mediate the axonal regenerative benefits remain largely unclear. Using the eye as the CNS model, here it is first indicated that placing mice in an exercise stimulation environment induced DNA methylation patterns and transcriptomes of retinal ganglion cell, promoted axon regeneration after injury, and reversed vision loss in aged mice. These beneficial effects are dependent on the DNA demethylases TET3-mediated epigenetic effects, which increased the expression of genes associated with the regenerative growth programs, such as STAT3, Wnt5a, Klf6. Exercise training also shows with the improved mitochondrial and metabolic dysfunction in retinas and optic nerves via TET3. Collectively, these results suggested that the increased regenerative capacity induced by enhancing physical activity is mediated through epigenetic reprogramming in mouse model of optic nerve injury and in aged mouse. Understanding the molecular mechanism underlying exercise-dependent neuronal plasticity led to the identification of novel targets for ameliorating pathologies associated with etiologically diverse diseases.
Collapse
Affiliation(s)
- Si Fang
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Yunxiang Ji
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Yilan Shen
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Simin Yang
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Hongli Zhang
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Wenfeng Xin
- College of Notoginseng Medicine and Pharmacy, Wenshan University, Wenshan, 663000, China
| | - Weidong Shi
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Wei Chen
- Multiscale Research Institute of Complex Systems, Department of Integrative Oncology in Fudan University Shanghai Cancer Center, Jingan District Central Hospital of Shanghai, Department of Otorhinolaryngology-Head and Neck Surgery in Huashan Hospital, Fudan University, Shanghai, 200433, China
| |
Collapse
|
23
|
Chrysafi P, Valenzuela-Vallejo L, Stefanakis K, Kelesidis T, Connelly MA, Mantzoros CS. Total and H-specific GDF-15 levels increase in caloric deprivation independently of leptin in humans. Nat Commun 2024; 15:5190. [PMID: 38890300 PMCID: PMC11189399 DOI: 10.1038/s41467-024-49366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Mitochondrial-secreted growth differentiation factor-15 (GDF-15) promotes weight loss in animals. Its effects in humans remain unclear, due to limited research and potential measurement interference from the H202D-variant. Our post-hoc analysis investigates total (irrespective of genetic variants) and H-specific GDF-15 (detected only in H202D-variant absence) in humans under acute and chronic energy deprivation, examining GDF-15 interaction with leptin (energy homeostasis regulator) and GDF-15 biologic activity modulation by the H202D-variant. Total and H-specific GDF-15 increased with acute starvation, and total GDF-15 increased with chronic energy deprivation, compared with healthy subjects and regardless of leptin repletion. Baseline GDF-15 positively correlated with triglyceride-rich particles and lipoproteins. During acute metabolic stress, GDF-15 associations with metabolites/lipids appeared to differ in subjects with the H202D-variant. Our findings suggest GDF-15 increases with energy deprivation in humans, questioning its proposed weight loss and suggesting its function as a mitokine, reflecting or mediating metabolic stress response.
Collapse
Affiliation(s)
- Pavlina Chrysafi
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Laura Valenzuela-Vallejo
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Konstantinos Stefanakis
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Theodoros Kelesidis
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 02215, USA
| | | | - Christos S Mantzoros
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
- Department of Medicine, Boston VA Healthcare System, Boston, MA, 90095, USA.
| |
Collapse
|
24
|
Sabaratnam R, Kristensen JM, Pedersen AJT, Kruse R, Handberg A, Wojtaszewski JFP, Højlund K. Acute Exercise Increases GDF15 and Unfolded Protein Response/Integrated Stress Response in Muscle in Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:1754-1764. [PMID: 38242693 DOI: 10.1210/clinem/dgae032] [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: 07/20/2023] [Revised: 12/27/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
CONTEXT Regular exercise is a key prevention strategy for obesity and type 2 diabetes (T2D). Exerkines secreted in response to exercise or recovery may contribute to improved systemic metabolism. Conversely, an impaired exerkine response to exercise and recovery may contribute to cardiometabolic diseases. OBJECTIVE We investigated if the exercise-induced regulation of the exerkine, growth differentiation factor 15 (GDF15) and its putative upstream regulators of the unfolded protein response (UPR)/integrated stress response (ISR) is impaired in skeletal muscle in patients with T2D compared with weight-matched glucose-tolerant men. METHODS Thirteen male patients with T2D and 14 age- and weight-matched overweight/obese glucose-tolerant men exercised at 70% of VO2max for 1 hour. Blood and skeletal muscle biopsies were sampled before, immediately after, and 3 hours into recovery. Serum and muscle transcript levels of GDF15 and key markers of UPR/ISR were determined. Additionally, protein/phosphorylation levels of key regulators in UPR/ISR were investigated. RESULTS Acute exercise increased muscle gene expression and serum GDF15 levels in both groups. In recovery, muscle expression of GDF15 decreased toward baseline, whereas serum GDF15 remained elevated. In both groups, acute exercise increased the expression of UPR/ISR markers, including ATF4, CHOP, EIF2K3 (encoding PERK), and PPP1R15A (encoding GADD34), of which only CHOP remained elevated 3 hours into recovery. Downstream molecules of the UPR/ISR including XBP1-U, XBP1-S, and EDEM1 were increased with exercise and 3 hours into recovery in both groups. The phosphorylation levels of eIF2α-Ser51, a common marker of unfolded protein response (UPR) and ISR, increased immediately after exercise in controls, but decreased 3 hours into recovery in both groups. CONCLUSION In conclusion, exercise-induced regulation of GDF15 and key markers of UPR/ISR are not compromised in patients with T2D compared with weight-matched controls.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Jonas M Kristensen
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Andreas J T Pedersen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Rikke Kruse
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, North Denmark Region, DK-9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark
| | - Jørgen F P Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Kurt Højlund
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense M, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| |
Collapse
|
25
|
Groarke JD, Crawford J, Collins SM, Lubaczewski SL, Breen DM, Harrington MA, Jacobs I, Qiu R, Revkin J, Rossulek MI, Saxena AR. Phase 2 study of the efficacy and safety of ponsegromab in patients with cancer cachexia: PROACC-1 study design. J Cachexia Sarcopenia Muscle 2024; 15:1054-1061. [PMID: 38500292 PMCID: PMC11154777 DOI: 10.1002/jcsm.13435] [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: 08/31/2023] [Revised: 12/06/2023] [Accepted: 12/27/2023] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Cancer cachexia is a multifactorial metabolic wasting syndrome characterized by anorexia, unintentional loss of weight involving both skeletal muscle and adipose tissues, progressive functional impairment and reduced survival. Therapeutic strategies for this serious condition are very limited. Growth differentiation factor 15 (GDF-15) is a cytokine that is implicated in cancer cachexia and may represent both a biomarker of cancer cachexia and a potential therapeutic target. Ponsegromab is a potent and selective humanized monoclonal antibody that inhibits GDF-15-mediated signalling. Preclinical and preliminary phase 1 data suggest that ponsegromab-mediated inactivation of circulating GDF-15 may lead to improvement in key characteristics of cachexia. The primary objective of this phase 2 study is to assess the effect of ponsegromab on body weight in patients with cancer, cachexia and elevated GDF-15 concentrations. Secondary objectives include assessing physical activity, physical function, actigraphy, appetite, nausea and vomiting, fatigue and safety. Exploratory objectives include evaluating pharmacokinetics, pharmacodynamics, immunogenicity, lumbar skeletal muscle index and Response Evaluation Criteria in Solid Tumors. METHODS Approximately 168 adults with non-small-cell lung, pancreatic or colorectal cancers who have cachexia and elevated GDF-15 concentrations will be randomized in a double-blind, placebo-controlled study (NCT05546476). Participants meeting eligibility criteria will be randomized 1:1:1:1 to one of three dose groups of ponsegromab (100, 200 or 400 mg) or matching placebo administered subcutaneously every 4 weeks for an initial 12-week treatment period. This is followed by optional open-label treatment with ponsegromab of 400 mg administered every 4 weeks for up to 1 year. The primary endpoint is mean change from baseline in body weight at Week 12. A mixed model for repeated measures followed by a Bayesian Emax model will be used for the primary analysis. Secondary endpoints include physical activity, physical function and actigraphy measured by remote digital sensors; patient-reported appetite-related symptoms assessed by Functional Assessment of Anorexia-Cachexia Therapy subscale scores; anorexia/appetite, nausea and vomiting, and fatigue evaluated according to questions from the Cancer-Related Cachexia Symptom Diary; and incidence of adverse events, safety laboratory tests, vital signs and electrocardiogram abnormalities. PERSPECTIVE Cancer-related cachexia is an area of significant unmet medical need. This study will support the clinical development of ponsegromab as a novel inhibitor of GDF-15, which may ameliorate key pathologies of cancer cachexia to improve patient symptoms, functionality and quality of life. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT05546476.
Collapse
Affiliation(s)
| | | | - Susie M. Collins
- Global Biometrics and Data ManagementPfizer R&D UK LtdSandwichKentUK
| | - Shannon L. Lubaczewski
- Early Clinical Development and Biomedicine Artificial IntelligencePfizer IncCollegevillePAUSA
| | | | | | - Ira Jacobs
- Global Product DevelopmentPfizer IncNew YorkNYUSA
| | - Ruolun Qiu
- Clinical PharmacologyPfizer IncCambridgeMAUSA
| | - James Revkin
- Internal Medicine Research UnitPfizer IncCambridgeMAUSA
| | | | | |
Collapse
|
26
|
Walzik D, Wences Chirino TY, Zimmer P, Joisten N. Molecular insights of exercise therapy in disease prevention and treatment. Signal Transduct Target Ther 2024; 9:138. [PMID: 38806473 PMCID: PMC11133400 DOI: 10.1038/s41392-024-01841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Despite substantial evidence emphasizing the pleiotropic benefits of exercise for the prevention and treatment of various diseases, the underlying biological mechanisms have not been fully elucidated. Several exercise benefits have been attributed to signaling molecules that are released in response to exercise by different tissues such as skeletal muscle, cardiac muscle, adipose, and liver tissue. These signaling molecules, which are collectively termed exerkines, form a heterogenous group of bioactive substances, mediating inter-organ crosstalk as well as structural and functional tissue adaption. Numerous scientific endeavors have focused on identifying and characterizing new biological mediators with such properties. Additionally, some investigations have focused on the molecular targets of exerkines and the cellular signaling cascades that trigger adaption processes. A detailed understanding of the tissue-specific downstream effects of exerkines is crucial to harness the health-related benefits mediated by exercise and improve targeted exercise programs in health and disease. Herein, we review the current in vivo evidence on exerkine-induced signal transduction across multiple target tissues and highlight the preventive and therapeutic value of exerkine signaling in various diseases. By emphasizing different aspects of exerkine research, we provide a comprehensive overview of (i) the molecular underpinnings of exerkine secretion, (ii) the receptor-dependent and receptor-independent signaling cascades mediating tissue adaption, and (iii) the clinical implications of these mechanisms in disease prevention and treatment.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Tiffany Y Wences Chirino
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, 37075, Göttingen, Lower Saxony, Germany.
| |
Collapse
|
27
|
Mulcahy MC, El Habbal N, Redd JR, Sun H, Gregg BE, Bridges D. GDF15 knockout does not substantially impact perinatal body weight or neonatal outcomes in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591359. [PMID: 38746399 PMCID: PMC11092610 DOI: 10.1101/2024.04.30.591359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Growth differentiation factor-15 (GDF15) increases in circulation during pregnancy and has been implicated in food intake, weight loss, complications of pregnancy, and metabolic illness. We used a Gdf15 knockout mouse model (Gdf15-/- ) to assess the role of GDF15 in body weight regulation and food intake during pregnancy. We found that Gdf15-/- dams consumed a similar amount of food and gained comparable weight during the course of pregnancy compared to Gdf15+/+ dams. Insulin sensitivity on gestational day 16.5 was also similar between genotypes. In the postnatal period, litter size, and survival rates were similar between genotypes. There was a modest reduction in birth weight of Gdf15-/- pups, but this difference was no longer evident postnatal day 3.5 to 14.5. We observed no detectable differences in milk volume production or milk fat percentage. These data suggest that GDF15 is dispensable for changes in food intake, and body weight as well as insulin sensitivity during pregnancy in a mouse model.
Collapse
Affiliation(s)
- Molly C. Mulcahy
- University of Michigan School of Public Health, Department of Nutritional Sciences
| | - Noura El Habbal
- University of Michigan School of Public Health, Department of Nutritional Sciences
| | - JeAnna R. Redd
- University of Michigan School of Public Health, Department of Nutritional Sciences
| | - Haijing Sun
- Michigan Medicine, Department of Pediatric Endocrinology
| | - Brigid E. Gregg
- University of Michigan School of Public Health, Department of Nutritional Sciences
- Michigan Medicine, Department of Pediatric Endocrinology
| | - Dave Bridges
- University of Michigan School of Public Health, Department of Nutritional Sciences
| |
Collapse
|
28
|
Pena-Leon V, Perez-Lois R, Villalon M, Folgueira C, Barja-Fernández S, Prida E, Baltar J, Santos F, Fernø J, García-Caballero T, Nogueiras R, Quiñones M, Al-Massadi O, Seoane LM. Gastric GDF15 levels are regulated by age, sex, and nutritional status in rodents and humans. J Endocrinol Invest 2024; 47:1139-1154. [PMID: 37955834 DOI: 10.1007/s40618-023-02232-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/21/2023] [Indexed: 11/14/2023]
Abstract
AIM Growth differentiation factor 15 (GDF15) is a stress response cytokine that has been proposed as a relevant metabolic hormone. Descriptive studies have shown that plasma GDF15 levels are regulated by short term changes in nutritional status, such as fasting, or in obesity. However, few data exist regarding how GDF15 levels are regulated in peripheral tissues. The aim of the present work was to study the variations on gastric levels of GDF15 and its precursor under different physiological conditions, such as short-term changes in nutritional status or overfeeding achieved by HFD. Moreover, we also address the sex- and age-dependent alterations in GDF15 physiology. METHODS The levels of gastric and plasma GDF15 and its precursor were measured in lean and obese mice, rats and humans by western blot, RT-PCR, ELISA, immunohistochemistry and by an in vitro organ culture system. RESULTS Our results show a robust regulation of gastric GDF15 production by fasting in rodents. In obesity an increase in GDF15 secretion from the stomach is reflected with an increase in circulating levels of GDF15 in rats and humans. Moreover, gastric GDF15 levels increase with age in both rats and humans. Finally, gastric GDF15 levels display sexual dimorphism, which could explain the difference in circulating GFD15 levels between males and females, observed in both humans and rodents. CONCLUSIONS Our results provide clear evidence that gastric GDF15 is a critical contributor of circulating GDF15 levels and can explain some of the metabolic effects induced by GDF15.
Collapse
Affiliation(s)
- V Pena-Leon
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - R Perez-Lois
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - M Villalon
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - C Folgueira
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - S Barja-Fernández
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - E Prida
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (IDIS/CHUS), Santiago de Compostela, Spain
| | - J Baltar
- Servicio de Cirugía General y del Aparato Digestivo, CHUS7SERGAS Santiago de Compostela, Rua R Baltar s/n, 15706, Santiago de Compostela, Spain
| | - F Santos
- Servicio de Cirugía General y del Aparato Digestivo, CHUS7SERGAS Santiago de Compostela, Rua R Baltar s/n, 15706, Santiago de Compostela, Spain
| | - J Fernø
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, 5201, Bergen, Norway
| | - T García-Caballero
- Departamento de Ciencias Morfologicas, Facultad de Medicina, USC, Complejo Hospitalario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - R Nogueiras
- Departamento de Fisiología, Instituto de Investigación Sanitaria de Santiago de Compostela, CIMUS, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - M Quiñones
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - O Al-Massadi
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (IDIS/CHUS), Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
| | - L M Seoane
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
| |
Collapse
|
29
|
Petersen J, Ludwig MQ, Juozaityte V, Ranea-Robles P, Svendsen C, Hwang E, Kristensen AW, Fadahunsi N, Lund J, Breum AW, Mathiesen CV, Sachs L, Moreno-Justicia R, Rohlfs R, Ford JC, Douros JD, Finan B, Portillo B, Grose K, Petersen JE, Trauelsen M, Feuchtinger A, DiMarchi RD, Schwartz TW, Deshmukh AS, Thomsen MB, Kohlmeier KA, Williams KW, Pers TH, Frølund B, Strømgaard K, Klein AB, Clemmensen C. GLP-1-directed NMDA receptor antagonism for obesity treatment. Nature 2024; 629:1133-1141. [PMID: 38750368 PMCID: PMC11136670 DOI: 10.1038/s41586-024-07419-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/12/2024] [Indexed: 05/25/2024]
Abstract
The N-methyl-D-aspartate (NMDA) receptor is a glutamate-activated cation channel that is critical to many processes in the brain. Genome-wide association studies suggest that glutamatergic neurotransmission and NMDA receptor-mediated synaptic plasticity are important for body weight homeostasis1. Here we report the engineering and preclinical development of a bimodal molecule that integrates NMDA receptor antagonism with glucagon-like peptide-1 (GLP-1) receptor agonism to effectively reverse obesity, hyperglycaemia and dyslipidaemia in rodent models of metabolic disease. GLP-1-directed delivery of the NMDA receptor antagonist MK-801 affects neuroplasticity in the hypothalamus and brainstem. Importantly, targeting of MK-801 to GLP-1 receptor-expressing brain regions circumvents adverse physiological and behavioural effects associated with MK-801 monotherapy. In summary, our approach demonstrates the feasibility of using peptide-mediated targeting to achieve cell-specific ionotropic receptor modulation and highlights the therapeutic potential of unimolecular mixed GLP-1 receptor agonism and NMDA receptor antagonism for safe and effective obesity treatment.
Collapse
Affiliation(s)
- Jonas Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Q Ludwig
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vaida Juozaityte
- 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
| | - Charlotte Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Eunsang Hwang
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Amalie W Kristensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicole Fadahunsi
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alberte W Breum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie V Mathiesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Luisa Sachs
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Roger Moreno-Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca Rohlfs
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - James C Ford
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | | | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Bryan Portillo
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kyle Grose
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Jacob E Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Trauelsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Annette Feuchtinger
- Core Facility Pathology & Tissue Analytics, Helmholtz Munich, Neuherberg, Germany
| | | | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kevin W Williams
- Center for Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Frølund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders B Klein
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
30
|
Steinmetz J, Stemmler A, Hennig CL, Symmank J, Jacobs C. GDF15 Contributes to the Regulation of the Mechanosensitive Responses of PdL Fibroblasts through the Modulation of IL-37. Dent J (Basel) 2024; 12:39. [PMID: 38392243 PMCID: PMC10888100 DOI: 10.3390/dj12020039] [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: 11/30/2023] [Revised: 01/28/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
During orthodontic tooth movement (OTM), areas of compressive and tensile forces are generated in the periodontal ligament (PdL), a mechanoreactive connective tissue between the teeth and alveolar bone. Mechanically stimulated PdL fibroblasts (PdLFs), the main cell type of PdL, express significantly increased levels of growth differentiation factor 15 (GDF15). In compressed PdL areas, GDF15 plays a fundamental role in modulating relevant OTM processes, including inflammation and osteoclast activation. However, the specific function of this factor in tensile areas has not yet been investigated. Thus, the aim of this study was to investigate the role of GDF15 in the mechanoresponse of human PdLFs (hPdLFs) that were exposed to biaxial tensile forces in vitro. Using siRNA-mediated knockdown experiments, we demonstrated that GDF15 had no impact on the anti-inflammatory force response of elongated hPdLFs. Although the anti-inflammatory markers IL1RN and IL10, as well as the activation of immune cells remained unaffected, we demonstrated an inhibitory role of GDF15 for the IL-37 expression. By analyzing osteogenic markers, including ALPL and RUNX2, along with an assessment of alkaline phosphatase activation, we further showed that the regulation of IL-37 by GDF15 modulates the osteogenic differentiation potential of hPdLFs. Despite bone resorption in tensile areas being rather limited, GDF15 was also found to positively modulate osteoclast activation in those areas, potentially by adjusting the IL-37 levels. In light of our new findings, we hypothesize that GDF15 modulates force-induced processes in tissue and bone remodeling through its various intra- and extracellular signaling pathways as well as interaction partners. Potentially acting as a master regulator, the modulation of GDF15 levels may hold relevance for clinical implications.
Collapse
Affiliation(s)
- Julia Steinmetz
- Department of Orthodontics, University Hospital Jena, 07743 Jena, Germany
| | - Albert Stemmler
- Department of Orthodontics, University Hospital Jena, 07743 Jena, Germany
| | | | - Judit Symmank
- Department of Orthodontics, University Hospital Jena, 07743 Jena, Germany
| | - Collin Jacobs
- Department of Orthodontics, University Hospital Jena, 07743 Jena, Germany
| |
Collapse
|
31
|
Lund C, Ranea-Robles P, Falk S, Rausch DM, Skovbjerg G, Vibe-Petersen VK, Krauth N, Skytte JL, Vana V, Roostalu U, Pers TH, Lund J, Clemmensen C. Protection against overfeeding-induced weight gain is preserved in obesity but does not require FGF21 or MC4R. Nat Commun 2024; 15:1192. [PMID: 38331907 PMCID: PMC10853283 DOI: 10.1038/s41467-024-45223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Overfeeding triggers homeostatic compensatory mechanisms that counteract weight gain. Here, we show that both lean and diet-induced obese (DIO) male mice exhibit a potent and prolonged inhibition of voluntary food intake following overfeeding-induced weight gain. We reveal that FGF21 is dispensable for this defense against weight gain. Targeted proteomics unveiled novel circulating factors linked to overfeeding, including the protease legumain (LGMN). Administration of recombinant LGMN lowers body weight and food intake in DIO mice. The protection against weight gain is also associated with reduced vascularization in the hypothalamus and sustained reductions in the expression of the orexigenic neuropeptide genes, Npy and Agrp, suggesting a role for hypothalamic signaling in this homeostatic recovery from overfeeding. Overfeeding of melanocortin 4 receptor (MC4R) KO mice shows that these mice can suppress voluntary food intake and counteract the enforced weight gain, although their rate of weight recovery is impaired. Collectively, these findings demonstrate that the defense against overfeeding-induced weight gain remains intact in obesity and involves mechanisms independent of both FGF21 and MC4R.
Collapse
Affiliation(s)
- Camilla Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Ranea-Robles
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Falk
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Dylan M Rausch
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Grethe Skovbjerg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Gubra ApS, Hørsholm, Denmark
| | | | - Nathalie Krauth
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Vasiliki Vana
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
32
|
Reyes J, Zhao Y, Pandya K, Yap GS. Growth differentiation factor-15 is an IFN-γ regulated mediator of infection-induced weight loss and the hepatic FGF21 response. Brain Behav Immun 2024; 116:24-33. [PMID: 38013040 DOI: 10.1016/j.bbi.2023.11.029] [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: 08/24/2023] [Revised: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 11/29/2023] Open
Abstract
Infections are often accompanied by weight loss caused by alterations in host behavior and metabolism, also known as sickness behaviors. Recent studies have revealed that sickness behaviors can either promote or impede survival during infections depending on factors such as the type of infectious pathogen. Nevertheless, we have an incomplete understanding of the underlying mechanisms of sickness behaviors. Furthermore, although the host immune responses to infections have long been known to contribute to the induction of sickness behaviors, recent studies have identified emerging cytokines that are also key regulators of host metabolism during infection and inflammation, such as growth differentiation factor 15 (GDF-15). GDF-15 is a distant member of the TGF-β superfamily that causes weight loss by suppressing appetite and food consumption and causing emesis. These effects require activation of neurons that express the only known GDF-15 receptor, the GFRAL receptor. GDF-15 also functions in the periphery including the induction of ketogenesis and immunoregulation. Nevertheless, the functions and regulation of GDF-15 during live infections is not yet known. Murine infection with avirulent Toxoplasma gondii is an established model to understand infection-induced weight loss. Past studies have determined that acute T. gondii infection causes weight loss due to diminished food consumption and increased energy expenditure through unknown mechanisms. Additionally, our lab previously demonstrated that T. gondii causes upregulation in serum GDF-15 in an IFN-γ-dependent manner during the post-acute phase of the infection. In this study, we interrogated the in-vivo functions and immune regulation of GDF-15 during Toxoplasma gondii infection. First, we found that in wild-type mice, acute T. gondii infection caused a significant weight loss that is preceded by elevation of serum levels of IFN-γ and GDF-15. To determine whether IFN-γ regulates GDF-15, we neutralized IFN-γ on days 5 and 6 and measured GDF-15 on day 7 and found that serum but not tissue levels of GDF-15 decreased after IFN-γ neutralization. Additionally, exogenous IFN-γ was sufficient to elevate serum GDF-15 in the absence of infection. Next, we compared the outcomes of T. gondii infection between WT and Gdf15-/- mice. We observed that the weight trajectories were declining in WT mice while they were increasing in Gdf15-/-mice during the acute phase of the infection. This difference in trajectories extended throughout the chronic infection resulting to an overall weight loss relative to initial weights in WT mice but not Gdf15-/-mice. Then, we determined that GDF-15 is not essential for survival and immunoregulation during T. gondii infection. We also demonstrated that GDF-15 is required for the induction of FGF21, stress-induced cytokine with prominent roles in regulating host metabolism. Finally, we discovered a cytokine cascade IFN-γ-GDF-15-FGF21 that is likely involved in the regulation of host metabolism. Overall, our study provides evidence that IFN-γ contributes to the regulation of host metabolism during infection by inducing GDF-15 and FGF21. GDF-15 orchestrates changes in host metabolism that supports the host immune response in clearing the infection. These physiological alterations induce FGF21, which in turn, orchestrates the adaptive responses to the effects of GDF-15, which can be detrimental when protracted.
Collapse
Affiliation(s)
- Jojo Reyes
- Department of Medicine and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Yanlin Zhao
- Department of Medicine and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Krushang Pandya
- Department of Medicine and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States; Program of Bioengineering, Department of Electrical & Computer Engineering, New York Institute of Technology, United States
| | - George S Yap
- Department of Medicine and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States.
| |
Collapse
|
33
|
Robberechts R, Poffé C. Defining ketone supplementation: the evolving evidence for postexercise ketone supplementation to improve recovery and adaptation to exercise. Am J Physiol Cell Physiol 2024; 326:C143-C160. [PMID: 37982172 DOI: 10.1152/ajpcell.00485.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023]
Abstract
Over the last decade, there has been a growing interest in the use of ketone supplements to improve athletic performance. These ketone supplements transiently elevate the concentrations of the ketone bodies acetoacetate (AcAc) and d-β-hydroxybutyrate (βHB) in the circulation. Early studies showed that ketone bodies can improve energetic efficiency in striated muscle compared with glucose oxidation and induce a glycogen-sparing effect during exercise. As such, most research has focused on the potential of ketone supplementation to improve athletic performance via ingestion of ketones immediately before or during exercise. However, subsequent studies generally observed no performance improvement, and particularly not under conditions that are relevant for most athletes. However, more and more studies are reporting beneficial effects when ketones are ingested after exercise. As such, the real potential of ketone supplementation may rather be in their ability to enhance postexercise recovery and training adaptations. For instance, recent studies observed that postexercise ketone supplementation (PEKS) blunts the development of overtraining symptoms, and improves sleep, muscle anabolic signaling, circulating erythropoietin levels, and skeletal muscle angiogenesis. In this review, we provide an overview of the current state-of-the-art about the impact of PEKS on aspects of exercise recovery and training adaptation, which is not only relevant for athletes but also in multiple clinical conditions. In addition, we highlight the underlying mechanisms by which PEKS may improve exercise recovery and training adaptation. This includes epigenetic effects, signaling via receptors, modulation of neurotransmitters, energy metabolism, and oxidative and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Ruben Robberechts
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Chiel Poffé
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
34
|
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
|
35
|
Nayak N, Mukherjee T, Pattnaik A. Comprehensive Role of GDF15 in Inhibiting Adipogenesis and Hyperlipidemia, Enhancing Cardiovascular Health and Alleviating Inflammation in Metabolic Disorders. Curr Pharm Des 2024; 30:2387-2399. [PMID: 38934286 DOI: 10.2174/0113816128318741240611114448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024]
Abstract
Growth Differentiation Factor 15 (GDF15) has emerged as a pivotal signaling molecule implicated in diverse physiological processes, spanning metabolic regulation, inflammation, and cardiovascular health. This study provides a comprehensive exploration of GDF15's multifaceted role, primarily focusing on its association with obesity-related complications and therapeutic potential. GDF15's involvement in energy homeostasis, specifically its regulation of body weight and appetite through hindbrain neuron activation and the GFRAL-RET signaling pathway, underscores its significance as an appetite-regulating hormone. GDF15's intricate modulation within adipose tissue dynamics in response to dietary changes and obesity, coupled with its influence on insulin sensitivity, highlights its critical role in metabolic health. The manuscript delves into the intricate crosstalk between GDF15 and pathways related to insulin sensitivity, macrophage polarization, and adipose tissue function, elucidating its potential as a therapeutic target for metabolic disorders associated with obesity. GDF15's association with chronic low-grade inflammation and its impact on cardiovascular health, particularly during hyperlipidemia and ischemic events, are explored. The intricate relationship between GDF15 and cardiovascular diseases, including its effects on endothelial function, cardiac hypertrophy, and heart failure, emphasizes its multifaceted nature in maintaining overall cardiovascular well-being. Challenges regarding the therapeutic application of GDF15, such as long-term safety concerns and ongoing clinical investigations, are discussed. Lastly, future research directions exploring GDF15's potential in addressing obesity-related complications and cardiovascular risks are proposed, highlighting its promising role as a therapeutic target in reshaping treatment strategies for obesity and associated health conditions.
Collapse
Affiliation(s)
- Nikita Nayak
- Department of Pharmaceutical Sciences and Technology, Division of Pharmacology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Tuhin Mukherjee
- Department of Pharmaceutical Sciences and Technology, Division of Pharmacology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Ashok Pattnaik
- Department of Pharmaceutical Sciences and Technology, Division of Pharmacology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| |
Collapse
|
36
|
Jia S, Yu Z, Bai L. Exerkines and osteoarthritis. Front Physiol 2023; 14:1302769. [PMID: 38107476 PMCID: PMC10722202 DOI: 10.3389/fphys.2023.1302769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease, with physical exercise being a widely endorsed strategy in its management guidelines. Exerkines, defined as cytokines secreted in response to acute and chronic exercise, function through endocrine, paracrine, and/or autocrine pathways. Various tissue-specific exerkines, encompassing exercise-induced myokines (muscle), cardiokines (heart), and adipokines (adipose tissue), have been linked to exercise therapy in OA. Exerkines are derived from these kines, but unlike them, only kines regulated by exercise can be called exerkines. Some of these exerkines serve a therapeutic role in OA, such as irisin, metrnl, lactate, secreted frizzled-related protein (SFRP), neuregulin, and adiponectin. While others may exacerbate the condition, such as IL-6, IL-7, IL-15, IL-33, myostatin, fractalkine, follistatin-like 1 (FSTL1), visfatin, activin A, migration inhibitory factor (MIF), apelin and growth differentiation factor (GDF)-15. They exerts anti-/pro-apoptosis/pyroptosis/inflammation, chondrogenic differentiation and cell senescence effect in chondrocyte, synoviocyte and mesenchymal stem cell. The modulation of adipokine effects on diverse cell types within the intra-articular joint emerges as a promising avenue for future OA interventions. This paper reviews recent findings that underscore the significant role of tissue-specific exerkines in OA, delving into the underlying cellular and molecular mechanisms involved.
Collapse
Affiliation(s)
- Shuangshuo Jia
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyao Yu
- Imaging Department, Dalian Medical University, Dalian, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
37
|
Jia D, Tian Z, Wang R. Exercise mitigates age-related metabolic diseases by improving mitochondrial dysfunction. Ageing Res Rev 2023; 91:102087. [PMID: 37832607 DOI: 10.1016/j.arr.2023.102087] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
The benefits of regular physical activity are related to delaying and reversing the onset of ageing and age-related disorders, including cardiomyopathy, neurodegenerative diseases, cancer, obesity, diabetes, and fatty liver diseases. However, the molecular mechanisms of the benefits of exercise or physical activity on ageing and age-related disorders remain poorly understood. Mitochondrial dysfunction is implicated in the pathogenesis of ageing and age-related metabolic diseases. Mitochondrial health is an important mediator of cellular function. Therefore, exercise alleviates metabolic diseases in individuals with advancing ageing and age-related diseases by the remarkable promotion of mitochondrial biogenesis and function. Exerkines are identified as signaling moieties released in response to exercise. Exerkines released by exercise have potential roles in improving mitochondrial dysfunction in response to age-related disorders. This review comprehensive summarizes the benefits of exercise in metabolic diseases, linking mitochondrial dysfunction to the onset of age-related diseases. Using relevant examples utilizing this approach, the possibility of designing therapeutic interventions based on these molecular mechanisms is addressed.
Collapse
Affiliation(s)
- Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
38
|
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
|
39
|
Lund J, Clemmensen C. Physiological protection against weight gain: evidence from overfeeding studies and future directions. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220229. [PMID: 37482786 PMCID: PMC10363696 DOI: 10.1098/rstb.2022.0229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/24/2023] [Indexed: 07/25/2023] Open
Abstract
Body weight is under physiological regulation. When body fat mass decreases, a series of responses are triggered to promote weight regain by increasing food intake and decreasing energy expenditure. Analogous, in response to experimental overfeeding, excessive weight gain is counteracted by a reduction in food intake and possibly by an increase in energy expenditure. While low blood leptin and other hormones defend against weight loss, the signals that oppose overfeeding-induced fat mass expansion are still unknown. In this article, we discuss insights gained from overfeeding interventions in humans and intragastric overfeeding studies in rodents. We summarize the knowledge on the relative contributions of energy intake, energy expenditure and energy excretion to the physiological defence against overfeeding-induced weight gain. Furthermore, we explore literature supporting the existence of unidentified endocrine and non-endocrine pathways that defend against weight gain. Finally, we discuss the physiological drivers of constitutional thinness and suggest that overfeeding of individuals with constitutional thinness represents a gateway to understand the physiology of weight gain resistance in humans. Experimental overfeeding, combined with modern multi-omics techniques, has the potential to unveil the long-sought signalling pathways that protect against weight gain. Discovering these mechanisms could give rise to new treatments for obesity. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research. Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research. Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
40
|
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: 9.0] [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
|
41
|
Zhang H, Mulya A, Nieuwoudt S, Vandanmagsar B, McDowell R, Heintz EC, Zunica ER, Collier JJ, Bozadjieva-Kramer N, Seeley RJ, Axelrod CL, Kirwan JP. GDF15 Mediates the Effect of Skeletal Muscle Contraction on Glucose-Stimulated Insulin Secretion. Diabetes 2023; 72:1070-1082. [PMID: 37224335 PMCID: PMC10382648 DOI: 10.2337/db22-0019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Exercise is a first-line treatment for type 2 diabetes and preserves β-cell function by hitherto unknown mechanisms. We postulated that proteins from contracting skeletal muscle may act as cellular signals to regulate pancreatic β-cell function. We used electric pulse stimulation (EPS) to induce contraction in C2C12 myotubes and found that treatment of β-cells with EPS-conditioned medium enhanced glucose-stimulated insulin secretion (GSIS). Transcriptomics and subsequent targeted validation revealed growth differentiation factor 15 (GDF15) as a central component of the skeletal muscle secretome. Exposure to recombinant GDF15 enhanced GSIS in cells, islets, and mice. GDF15 enhanced GSIS by upregulating the insulin secretion pathway in β-cells, which was abrogated in the presence of a GDF15 neutralizing antibody. The effect of GDF15 on GSIS was also observed in islets from GFRAL-deficient mice. Circulating GDF15 was incrementally elevated in patients with pre- and type 2 diabetes and positively associated with C-peptide in humans with overweight or obesity. Six weeks of high-intensity exercise training increased circulating GDF15 concentrations, which positively correlated with improvements in β-cell function in patients with type 2 diabetes. Taken together, GDF15 can function as a contraction-induced protein that enhances GSIS through activating the canonical signaling pathway in a GFRAL-independent manner. ARTICLE HIGHLIGHTS Exercise improves glucose-stimulated insulin secretion through direct interorgan communication. Contracting skeletal muscle releases growth differentiation factor 15 (GDF15), which is required to synergistically enhance glucose-stimulated insulin secretion. GDF15 enhances glucose-stimulated insulin secretion by activating the canonical insulin release pathway. Increased levels of circulating GDF15 after exercise training are related to improvements in β-cell function in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Anny Mulya
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Stephan Nieuwoudt
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Bolormaa Vandanmagsar
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Ruth McDowell
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Elizabeth C. Heintz
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Elizabeth R.M. Zunica
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - J. Jason Collier
- Islet Biology and Inflammation Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, Ann Arbor, MI
- Veterans Affairs Ann Arbor Healthcare System, Research Service, Ann Arbor, MI
| | - Randy J. Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Christopher L. Axelrod
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - John P. Kirwan
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
42
|
Karusheva Y, Petry CJ, Yasara N, Kottahachchi D, Premawardhena A, Barker P, Burling K, Sattar N, Welsh P, Mettananda S, O'Rahilly S. Association of GDF15 levels with body mass index and endocrine status in β-thalassaemia. Clin Endocrinol (Oxf) 2023; 99:182-189. [PMID: 36806122 PMCID: PMC10952638 DOI: 10.1111/cen.14897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Abstract
OBJECTIVE GDF15 has emerged as a stress-induced hormone, acting on the brain to reduce food intake and body weight while affecting neuroendocrine function. Very high GDF15 levels are found in thalassaemia, where growth, energy balance and neuroendocrine function are impaired. We examined the relationships between GDF15 and anthropometric measures and endocrine status in β-thalassaemia. DESIGN Cross sectional study. PATIENTS All β-thalassaemia patients attending the thalassaemia unit of Colombo North Teaching Hospital for blood transfusions. MEASUREMENTS Anthropometric data, appetite scores, circulating GDF15, IGF, thyroid and reproductive hormone levels in 103 β-thalassaemia patients were obtained. RESULTS GDF15 levels were markedly elevated in thalassaemia patients (24.2-fold with β-thalassaemia major compared with healthy controls). Among patients with β-thalassaemia major, the relationship between GDF15 and body mass index (BMI) was curvilinear with all individuals with GDF15 levels above 24,000 pg/mL having a BMI below 20 kg/m2 . After adjustment for BMI, age and Tanner stage, serum IGF1 concentrations correlated negatively with GDF15 in all thalassaemia patients (β = -.027, p = .02). We found a significant positive relationship between GDF15 and gonadotropin (in both sexes) and testosterone (in males). CONCLUSIONS GDF15 levels were markedly elevated in patients with β-thalassaemia and its association with BMI is consistent with the known effect of GDF15 to reduce body weight. The inverse association between GDF15 with IGF1 levels may reflect a neuroendocrine impact of GDF15 or an indirect effect via impaired nutritional state. The positive association with testosterone in males and gonadotropins in both sexes, was surprising and should prompt further GDF15 studies on the hypothalamic pituitary gonadal axis.
Collapse
Affiliation(s)
- Yanislava Karusheva
- MRC Metabolic Diseases Unit, Wellcome‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- NIHR Cambridge Biomedical Research CentreCambridgeUK
| | - Clive J. Petry
- MRC Metabolic Diseases Unit, Wellcome‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- NIHR Cambridge Biomedical Research CentreCambridgeUK
| | - Nirmani Yasara
- Department of Paediatrics, Faculty of MedicineUniversity of KelaniyaRagamaSri Lanka
| | - Dulani Kottahachchi
- Department of Physiology, Faculty of MedicineUniversity of KelaniyaRagamaSri Lanka
| | - Anuja Premawardhena
- Colombo North Teaching HospitalRagamaSri Lanka
- Department of Medicine, Faculty of MedicineUniversity of KelaniyaRagamaSri Lanka
| | - Peter Barker
- NIHR Cambridge Biomedical Research CentreCambridgeUK
- Core Biochemical Assay LaboratoryCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Keith Burling
- NIHR Cambridge Biomedical Research CentreCambridgeUK
- Core Biochemical Assay LaboratoryCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Naveed Sattar
- University of Glasgow, School of Cardiovascular and Metabolic HealthGlasgowUK
| | - Paul Welsh
- University of Glasgow, School of Cardiovascular and Metabolic HealthGlasgowUK
| | - Sachith Mettananda
- Department of Paediatrics, Faculty of MedicineUniversity of KelaniyaRagamaSri Lanka
- Colombo North Teaching HospitalRagamaSri Lanka
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Wellcome‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- NIHR Cambridge Biomedical Research CentreCambridgeUK
| |
Collapse
|
43
|
Małkowska P, Sawczuk M. Cytokines as Biomarkers for Evaluating Physical Exercise in Trained and Non-Trained Individuals: A Narrative Review. Int J Mol Sci 2023; 24:11156. [PMID: 37446334 DOI: 10.3390/ijms241311156] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Physical activity and exercise training have numerous health benefits, including the prevention and management of chronic diseases, improvement of cardiovascular health, and enhancement of mental well-being. However, the effectiveness of training programs can vary widely among individuals due to various factors, such as genetics, lifestyle, and environment. Thus, identifying reliable biomarkers to evaluate physical training effectiveness and personalize training programs is crucial. Cytokines are signaling molecules produced by immune cells that play a vital role in inflammation and tissue repair. In recent years, there has been increasing interest in the potential use of cytokines as biomarkers for evaluating training effectiveness. This review article aims to provide an overview of cytokines, their potential as biomarkers, methods for measuring cytokine levels, and factors that can affect cytokine levels. The article also discusses the potential benefits of using cytokines as biomarkers, such as monitoring muscle damage and inflammation, and the potential for personalized training programs based on cytokine responses. We believe that the use of cytokines as biomarkers holds great promise for optimizing training programs and improving overall health outcomes.
Collapse
Affiliation(s)
- Paulina Małkowska
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland
- Doctoral School, University of Szczecin, 70-384 Szczecin, Poland
| | - Marek Sawczuk
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland
| |
Collapse
|
44
|
Barroso E, Montori-Grau M, Wahli W, Palomer X, Vázquez-Carrera M. Striking a gut-liver balance for the antidiabetic effects of metformin. Trends Pharmacol Sci 2023; 44:457-473. [PMID: 37188578 DOI: 10.1016/j.tips.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023]
Abstract
Metformin is the most prescribed drug for the treatment of type 2 diabetes mellitus (T2DM), but its mechanism of action has not yet been completely elucidated. Classically, the liver has been considered the major site of action of metformin. However, over the past few years, advances have unveiled the gut as an additional important target of metformin, which contributes to its glucose-lowering effect through new mechanisms of action. A better understanding of the mechanistic details of metformin action in the gut and the liver and its relevance in patients remains the challenge of present and future research and may impact drug development for the treatment of T2DM. Here, we offer a critical analysis of the current status of metformin-driven multiorgan glucose-lowering effects.
Collapse
Affiliation(s)
- Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, E-08950 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Avinguda Joan XXII 27-31, E-08028 Barcelona, Spain
| | - Marta Montori-Grau
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, E-08950 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Avinguda Joan XXII 27-31, E-08028 Barcelona, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland; Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore; ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, 31300 Toulouse Cedex, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, E-08950 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Avinguda Joan XXII 27-31, E-08028 Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, E-08950 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Avinguda Joan XXII 27-31, E-08028 Barcelona, Spain.
| |
Collapse
|
45
|
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
|
46
|
Quist JS, Klein AB, Færch K, Beaulieu K, Rosenkilde M, Gram AS, Sjödin A, Torekov S, Stallknecht B, Clemmensen C, Blond MB. Effects of acute exercise and exercise training on plasma GDF15 concentrations and associations with appetite and cardiometabolic health in individuals with overweight or obesity - A secondary analysis of a randomized controlled trial. Appetite 2023; 182:106423. [PMID: 36563967 DOI: 10.1016/j.appet.2022.106423] [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: 07/27/2022] [Revised: 11/09/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Growth Differentiation Factor 15 (GDF15) is seemingly involved in appetite control. Acute exercise increases GDF15 concentrations in lean humans, but acute and long-term effects of exercise on GDF15 in individuals with overweight/obesity are unknown. We investigated the effects of acute exercise and exercise training on GDF15 concentrations in individuals with overweight/obesity and associations with appetite and cardiometabolic markers. 90 physically inactive adults (20-45 years) with overweight/obesity were randomized to 6-months habitual lifestyle (CON, n=16), or isocaloric exercise of moderate (MOD, n=37) or vigorous intensity (VIG, n=37), 5 days/week. Testing was performed at baseline, 3, and 6 months. Plasma GDF15 concentrations, other metabolic markers, and subjective appetite were assessed fasted and in response to acute exercise before an ad libitum meal. Cardiorespiratory fitness, body composition, insulin sensitivity, and intraabdominal adipose tissue were measured. At baseline, GDF15 increased 18% (95%CI: 4; 34) immediately after acute exercise and 32% (16; 50) 60 min post-exercise. Fasting GDF15 increased 21% (0; 46) in VIG after 3 months (p=0.045), but this attenuated at 6 months (13% (-11; 43), p=0.316) and was unchanged in MOD (11% (-6; 32), p=0.224, across 3 and 6 months). Post-exercise GDF15 did not change in MOD or VIG. GDF15 was not associated with appetite or energy intake. Higher GDF15 was associated with lower cardiorespiratory fitness, central obesity, dyslipidemia, and poorer glycemic control. In conclusion, GDF15 increased in response to acute exercise but was unaffected by exercise training. Higher GDF15 concentrations were associated with a less favorable cardiometabolic profile but not with markers of appetite. This suggests that GDF15 increases in response to acute exercise independent of training state. Whether this has an impact on free-living energy intake and body weight management needs investigation.
Collapse
Affiliation(s)
- Jonas Salling Quist
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark; Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, DK-2730, Herlev, Denmark; School of Psychology, Faculty of Medicine & Health, University of Leeds, Woodhouse Lane, West Yorkshire, LS2 9JT, Leeds, United Kingdom.
| | - Anders Bue Klein
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen N, Denmark
| | - Kristine Færch
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark; Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, DK-2730, Herlev, Denmark
| | - Kristine Beaulieu
- Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, DK-2730, Herlev, Denmark; School of Psychology, Faculty of Medicine & Health, University of Leeds, Woodhouse Lane, West Yorkshire, LS2 9JT, Leeds, United Kingdom
| | - Mads Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark
| | - Anne Sofie Gram
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark
| | - Anders Sjödin
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 26, DK-1958, Fredederiksberg C, Denmark
| | - Signe Torekov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark
| | - Bente Stallknecht
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, DK-2200, Copenhagen N, Denmark
| | - Martin Bæk Blond
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark; Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, DK-2730, Herlev, Denmark
| |
Collapse
|
47
|
Benichou O, Coskun T, Gonciarz MD, Garhyan P, Adams AC, Du Y, Dunbar JD, Martin JA, Mather KJ, Pickard RT, Reynolds VL, Robins DA, Zvada SP, Emmerson PJ. Discovery, development, and clinical proof of mechanism of LY3463251, a long-acting GDF15 receptor agonist. Cell Metab 2023; 35:274-286.e10. [PMID: 36630958 DOI: 10.1016/j.cmet.2022.12.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/29/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023]
Abstract
GDF15 and its receptor GFRAL/RET form a non-homeostatic system that regulates food intake and body weight in preclinical species. Here, we describe a GDF15 analog, LY3463251, a potent agonist at the GFRAL/RET receptor with prolonged pharmacokinetics. In rodents and obese non-human primates, LY3463251 decreased food intake and body weight with no signs of malaise or emesis. In a first-in-human study in healthy participants, single subcutaneous LY3463251 injections showed a safety and pharmacokinetic profile supporting further clinical development with dose-dependent nausea and emesis in a subset of individuals. A subsequent 12-week multiple ascending dose study in overweight and obese participants showed that LY3463251 induced significant decreases in food intake and appetite scores associated with modest body weight reduction independent of nausea and emesis (clinicaltrials.gov: NCT03764774). These observations demonstrate that agonism of the GFRAL/RET system can modulate energy balance in humans, though the decrease in body weight is surprisingly modest, suggesting challenges in leveraging the GDF15 system for clinical weight-loss applications.
Collapse
Affiliation(s)
| | - Tamer Coskun
- Eli Lilly and Company, Indianapolis, IN 46285, USA
| | | | | | | | - Yu Du
- Eli Lilly and Company, Indianapolis, IN 46285, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chelette B, Chidomere CL, Dantzer R. The GDF15-GFRAL axis mediates chemotherapy-induced fatigue in mice. Brain Behav Immun 2023; 108:45-54. [PMID: 36427806 PMCID: PMC9868083 DOI: 10.1016/j.bbi.2022.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer-related fatigue is defined as a distressing persistent subjective sense of physical, emotional, and/or cognitive tiredness or exhaustion related to cancer or cancer treatment that is not proportional to recent activity and that interferes with usual functioning. This form of fatigue is highly prevalent during cancer treatment and in some patients, it can persist for years after treatment has ended. An understanding of the mechanisms that drive cancer-related fatigue is still lacking, which hampers the identification of effective treatment options. Various chemotherapeutic agents including cisplatin are known to induce mitochondrial dysfunction and this effect is known to mediate chemotherapy-induced peripheral neuropathy and cognitive dysfunction. Mitochondrial dysfunction results in the release of mitokines that act locally and at distance to promote metabolic and behavioral adjustments to this form of cellular stress. One of these mitokines, growth differentiation factor 15 (GDF15) and its receptor, glial cell line-derived neurotrophic factor family receptor α-like (GFRAL), have received special attention in oncology as activation of GFRAL mediates the anorexic response that is responsible for cancer anorexia. The present study was initiated to determine whether GDF15 and GFRAL are involved in cisplatin-induced fatigue. We first tested the ability of cisplatin to increase circulating GDF15 in mice before assessing whether GDF15 can induce behavioral fatigue measured by decreased wheel running in healthy mice and increase behavioral fatigue induced by cisplatin. Mice administered a long acting form of GDF15, mGDF15-fc, decreased their voluntary wheel running activity. When the same treatment was administered to mice receiving cisplatin, it increased the amplitude and duration of cisplatin-induced decrease in wheel running. To determine whether endogenous GDF15 mediates the behavioral fatigue induced by cisplatin, we then administered a neutralizing monoclonal antibody to GFRAL to mice injected with cisplatin. The GFRAL neutralizing antibody mostly prevented cisplatin-induced decrease in wheel running and accelerated recovery. Taken together these findings demonstrate for the first time the role of the GDF15/GFRAL axis in cisplatin-induced behaviors and indicate that this axis could be a promising therapeutic target for the treatment of cancer-related fatigue.
Collapse
Affiliation(s)
- Brandon Chelette
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chinenye L Chidomere
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Dantzer
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
49
|
Ladang A, Beaudart C, Reginster JY, Al-Daghri N, Bruyère O, Burlet N, Cesari M, Cherubini A, da Silva MC, Cooper C, Cruz-Jentoft AJ, Landi F, Laslop A, Maggi S, Mobasheri A, Ormarsdottir S, Radermecker R, Visser M, Yerro MCP, Rizzoli R, Cavalier E. Biochemical Markers of Musculoskeletal Health and Aging to be Assessed in Clinical Trials of Drugs Aiming at the Treatment of Sarcopenia: Consensus Paper from an Expert Group Meeting Organized by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) and the Centre Académique de Recherche et d'Expérimentation en Santé (CARES SPRL), Under the Auspices of the World Health Organization Collaborating Center for the Epidemiology of Musculoskeletal Conditions and Aging. Calcif Tissue Int 2023; 112:197-217. [PMID: 36633611 PMCID: PMC9859913 DOI: 10.1007/s00223-022-01054-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/18/2022] [Indexed: 01/13/2023]
Abstract
In clinical trials, biochemical markers provide useful information on the drug's mode of action, therapeutic response and side effect monitoring and can act as surrogate endpoints. In pharmacological intervention development for sarcopenia management, there is an urgent need to identify biomarkers to measure in clinical trials and that could be used in the future in clinical practice. The objective of the current consensus paper is to provide a clear list of biochemical markers of musculoskeletal health and aging that can be recommended to be measured in Phase II and Phase III clinical trials evaluating new chemical entities for sarcopenia treatment. A working group of the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) proposed classifying biochemical markers into 2 series: biochemical markers evaluating musculoskeletal status and biochemical markers evaluating causal factors. For series 1, the group agreed on 4 biochemical markers that should be assessed in Phase II or Phase III trials (i.e., Myostatin-Follistatin, Brain Derived Neurotrophic Factor, N-terminal Type III Procollagen and Serum Creatinine to Serum Cystatin C Ratio - or the Sarcopenia Index). For series 2, the group agreed on 6 biochemical markers that should be assessed in Phase II trials (i.e., the hormones insulin-like growth factor-1 (IGF-I), dehydroepiandrosterone sulphate, and cortisol, and the inflammatory markers C-reactive protein (CRP), interleukin-6 and tumor necrosis factor-α), and 2 in Phase III trials (i.e., IGF-I and CRP). The group also proposed optional biochemical markers that may provide insights into the mode of action of pharmacological therapies. Further research and development of new methods for biochemical marker assays may lead to the evolution of these recommendations.
Collapse
Affiliation(s)
- Aurélie Ladang
- Department of Clinical Chemistry, CHU de Liège, University of Liège, Liège, Belgium.
| | - Charlotte Beaudart
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Jean-Yves Reginster
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
- Biochemistry Department, College of Science, Chair for Biomarkers of Chronic Diseases, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Nasser Al-Daghri
- Biochemistry Department, College of Science, Chair for Biomarkers of Chronic Diseases, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Olivier Bruyère
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Nansa Burlet
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonio Cherubini
- Geriatric Unit, IRCCS Istituti Clinici Scientifici Maugeri, Milan, Italy
| | | | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | | | - Francesco Landi
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart, Rome, Italy
| | - Andrea Laslop
- Scientific Office, Federal Office for Safety in Health Care, Vienna, Austria
| | | | - Ali Mobasheri
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | - Régis Radermecker
- Department of Diabetes, Nutrition and Metabolic Disorders, Clinical Pharmacology, University of Liege, CHU de Liège, Liège, Belgium
| | - Marjolein Visser
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - René Rizzoli
- Faculty of Medicine, Service of Bone Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Etienne Cavalier
- Department of Clinical Chemistry, CHU de Liège, University of Liège, Liège, Belgium
| |
Collapse
|
50
|
Kim-Muller JY, Song L, LaCarubba Paulhus B, Pashos E, Li X, Rinaldi A, Joaquim S, Stansfield JC, Zhang J, Robertson A, Pang J, Opsahl A, Boucher M, Breen D, Hales K, Sheikh A, Wu Z, Zhang BB. GDF15 neutralization restores muscle function and physical performance in a mouse model of cancer cachexia. Cell Rep 2023; 42:111947. [PMID: 36640326 DOI: 10.1016/j.celrep.2022.111947] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/06/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cancer cachexia is a disorder characterized by involuntary weight loss and impaired physical performance. Decline in physical performance of patients with cachexia is associated with poor quality of life, and currently there are no effective pharmacological interventions that restore physical performance. Here we examine the effect of GDF15 neutralization in a mouse model of cancer-induced cachexia (TOV21G) that manifests weight loss and muscle function impairments. With comprehensive assessments, our results demonstrate that cachectic mice treated with the anti-GDF15 antibody mAB2 exhibit body weight gain with near-complete restoration of muscle mass and markedly improved muscle function and physical performance. Mechanistically, the improvements induced by GDF15 neutralization are primarily attributed to increased caloric intake, while altered gene expression in cachectic muscles is restored in caloric-intake-dependent and -independent manners. The findings indicate potential of GDF15 neutralization as an effective therapy to enhance physical performance of patients with cachexia.
Collapse
Affiliation(s)
- Ja Young Kim-Muller
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - LouJin Song
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Brianna LaCarubba Paulhus
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Evanthia Pashos
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Xiangping Li
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Anthony Rinaldi
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Stephanie Joaquim
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - John C Stansfield
- Biostatistics, Early Clinical Development, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Jiangwei Zhang
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 10777 Science Center Dr., San Diego, CA, USA
| | - Andrew Robertson
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Jincheng Pang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Alan Opsahl
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Danna Breen
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Katherine Hales
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Abdul Sheikh
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Bei B Zhang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA.
| |
Collapse
|