1
|
Rose JP, Morgan DA, Sullivan AI, Fu X, Inigo-Vollmer M, Burgess SC, Meyerholz DK, Rahmouni K, Potthoff MJ. FGF21 reverses MASH through coordinated actions on the CNS and liver. Cell Metab 2025:S1550-4131(25)00252-9. [PMID: 40367940 DOI: 10.1016/j.cmet.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/24/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, metabolic dysfunction-associated steatohepatitis (MASH), represent a growing public health burden with limited therapeutic options. Recent studies have revealed that fibroblast growth factor 21 (FGF21)-based analogs can significantly improve MASH, but the mechanisms for this effect are not well understood. Here, we demonstrate that the beneficial metabolic effects of FGF21 to reverse MASH are mediated through distinct mechanisms to independently lower hepatic triglyceride and cholesterol levels. Specifically, FGF21 signaling directly to glutamatergic neurons in the central nervous system (CNS) stimulates hepatic triglyceride reduction and reversal of fibrosis, whereas FGF21 signaling directly to hepatocytes is necessary and sufficient to reduce hepatic cholesterol levels in mice. Mechanistically, we show that FGF21 acts in the CNS to increase sympathetic nerve activity to the liver, which suppresses hepatic de novo lipogenesis. These results provide critical insights into a promising pharmacological target to treat MASH.
Collapse
Affiliation(s)
- Jesse P Rose
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andrew I Sullivan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Melissa Inigo-Vollmer
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - David K Meyerholz
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Veterans' Affairs Medical Center, Iowa City, IA 52242, USA; Harold Hamm Diabetes Center, University of Oklahoma Health Sciences, Oklahoma City, OK 73117, USA.
| |
Collapse
|
2
|
Mia S, Siokatas G, Sidiropoulou R, Hoffman M, Fragkiadakis K, Markopoulou E, Elesawy MI, Roy R, Blair S, Kuwabara Y, Rapushi E, Chaudhuri D, Makarewich CA, Gao E, Koch WJ, Schilling JD, Molkentin JD, Marketou M, Drosatos K. Hepato-cardiac interorgan communication controls cardiac hypertrophy via combined endocrine-autocrine FGF21 signaling. Cell Rep Med 2025:102125. [PMID: 40339570 DOI: 10.1016/j.xcrm.2025.102125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/13/2025] [Accepted: 04/11/2025] [Indexed: 05/10/2025]
Abstract
Fibroblast growth factor (FGF) 21 is a hormone produced mainly by the liver but also other organs, including the heart. Although FGF21 analogs are used for treating obesity and metabolic syndrome in humans, preclinical and clinical studies have elicited mixed results about whether prolonged FGF21 signaling is protective or detrimental for cardiac function. Based on our findings, showing elevated serum and cardiac FGF21 levels in humans with increased left ventricular afterload, we explore the involvement of FGF21 in cardiac hypertrophy. Our mouse studies reveal interorgan liver-heart crosstalk, which is controlled by an initial hepatic FGF21 release followed by the induction of cardiomyocyte (CM) FGF21 expression. Tissue-specific genetic ablation or anti-sense oligonucleotide-based inhibition of FGF21 shows that, in response to pressure overload, CM FGF21 upregulation is a critical event that is stimulated by liver-derived FGF21 and drives cardiac hypertrophy likely by interfering with cardioprotective oxytocin signaling. Conclusively, the hepato-cardiac FGF21-based signaling axis governs cardiac hypertrophy.
Collapse
Affiliation(s)
- Sobuj Mia
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Georgios Siokatas
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Rafailia Sidiropoulou
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthew Hoffman
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | | | - Eftychia Markopoulou
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Mahmoud I Elesawy
- Department of Immunology and Pathology, Washington University in St. Louis, St. Louis, MO, USA; Divison of Cardiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Rajika Roy
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Scott Blair
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Yasuhide Kuwabara
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Erjola Rapushi
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Dipayan Chaudhuri
- Division of Cardiovascular Medicine, Department of Internal Medicine, Biochemistry, Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Catherine A Makarewich
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Erhe Gao
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Walter J Koch
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Joel D Schilling
- Department of Immunology and Pathology, Washington University in St. Louis, St. Louis, MO, USA; Divison of Cardiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Maria Marketou
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
3
|
Wedemeyer SA, Jones NE, Raza IGA, Green FM, Xiao Y, Semwal MK, Garza AK, Archuleta KS, Wimberly KL, Venables T, Holländer GA, Griffith AV. Paracrine FGF21 dynamically modulates mTOR signaling to regulate thymus function across the lifespan. NATURE AGING 2025; 5:588-606. [PMID: 39972173 PMCID: PMC12003089 DOI: 10.1038/s43587-024-00801-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/20/2024] [Indexed: 02/21/2025]
Abstract
Consequences of age-associated thymic atrophy include declining T-cell responsiveness to pathogens and vaccines and diminished T-cell self-tolerance. Cortical thymic epithelial cells (cTECs) are primary targets of thymic aging, and recent studies suggested that their maintenance requires mTOR signaling downstream of medullary TEC (mTEC)-derived growth factors. Here, to test this hypothesis, we generated a knock-in mouse model in which FGF21 and mCherry are expressed by most mTECs. We find that mTEC-derived FGF21 promotes temporally distinct patterns of mTORC1 and mTORC2 signaling in cTECs, promotes thymus and individual cTEC growth and maintenance, increases T-cell responsiveness to viral infection, and diminishes indicators of peripheral autoimmunity in older mice. The effects of FGF21 overexpression on thymus size and mTOR signaling were abrogated by treatment with the mTOR inhibitor rapamycin. These results reveal a mechanism by which paracrine FGF21 signaling regulates thymus size and function throughout the lifespan, as well as potential therapeutic targets for improving T-cell function and tolerance in aging.
Collapse
Affiliation(s)
- Sarah A Wedemeyer
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Nicholas E Jones
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Iwan G A Raza
- Medical Sciences Division, University of Oxford, Oxford, UK
| | - Freedom M Green
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Yangming Xiao
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Manpreet K Semwal
- Sam and Ann Barshop Institute for Aging and Longevity Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Math and Science, Our Lady of the Lake University, San Antonio, TX, USA
| | - Aaron K Garza
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Kahealani S Archuleta
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Kymberly L Wimberly
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Thomas Venables
- Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Georg A Holländer
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, UK
- Paediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital, Basel, Switzerland
- Developmental Immunology, Department of Biosystems and Engineering, ETH Zurich, Zurich, Switzerland
| | - Ann V Griffith
- Department of Microbiology, Immunology, & Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA.
- Sam and Ann Barshop Institute for Aging and Longevity Studies, UT Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
4
|
Raji A, Gantz I, Crutchlow M, Flynn H, Xu L, Rodgers AJ, Krishnan R, Rizk ML, Hu S, Kaufman KD, Engel SS. Clinical Trial: A Phase 2b Study to Evaluate the Efficacy and Safety of MK-3655 in Individuals With Pre-Cirrhotic MASH. Aliment Pharmacol Ther 2025; 61:1152-1162. [PMID: 39984821 PMCID: PMC11908112 DOI: 10.1111/apt.70038] [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: 11/26/2024] [Revised: 01/04/2025] [Accepted: 02/08/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) is a metabolic regulator with demonstrated efficacy for the treatment of metabolic dysfunction-associated steatohepatitis (MASH). FGF21 signals through 'c' isoforms of the FGF receptors (FGFR) 1-3 and the co-receptor β-klotho. AIMS We report the safety and efficacy of MK-3655, a monoclonal antibody that binds β-klotho and selectively activates the FGFR1c/β-klotho co-receptor complex, in patients with pre-cirrhotic MASH. METHODS Phase 2b, randomised, multicenter, double-blind, placebo-controlled, parallel-group study in patients with pre-cirrhotic MASH (NAS ≥ 4 and MASH CRN fibrosis score Stage 2 or 3). Participants were randomised 1:1:1:1 to receive MK-3655 50 mg, 100 mg, 300 mg, or matching placebo subcutaneously every 4 weeks. The primary endpoint was MASH resolution without worsening of fibrosis by histology at Week 52. An interim analysis (IA) of liver fat content (LFC) was planned once ≥ 25 participants per treatment group completed an MRI-PDFF assessment at Week 24. RESULTS Among 183 participants, mean BMI was 33.4 kg/m2, mean LFC was 18.1%, and 52.5% had type 2 diabetes. At the IA, the differences from placebo in relative reduction from baseline in LFC were assessed as insufficient for continuation of the trial. Among participants with Week 24 LFC assessment, percent relative reductions from baseline (LS mean difference vs. placebo) for MK-3655 50 mg (N = 33), 100 mg (N = 36), and 300 mg (N = 31), were 19.1%, 19.0%, and 26.1%, respectively. MK-3655 was generally well tolerated. CONCLUSIONS In patients with pre-cirrhotic MASH, treatment with MK-3655 resulted in a modest reduction in LFC at 24 weeks. CLINICAL TRIAL NUMBER EudraCT: 2019-003048-63; NCT: 04583423.
Collapse
Affiliation(s)
| | - Ira Gantz
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | | | | | | | | | | | - Shuai Hu
- Merck & Co., Inc.RahwayNew JerseyUSA
| | | | | | | |
Collapse
|
5
|
Chen L, Gao M, Ong SB, Gong G. Functions of FGF21 and its role in cardiac hypertrophy. J Adv Res 2025:S2090-1232(25)00148-1. [PMID: 40089060 DOI: 10.1016/j.jare.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND FGF21 is a stress-inducible hormone that operates in the autocrine or paracrine manner. Recent reports have revealed that FGF21 is highly expressed in cardiac hypertrophy to protect against heart injury and dysfunction. FGF21 is used to treat cardiac hypertrophy in mouse models. However, preclinical and clinical trials are restricted. AIM OF REVIEW This review mainly elucidates the diverse functions of FGF21 and explores the relationship between these functions and cardiac hypertrophy. It also discusses challenges and future perspectives in treating cardiac hypertrophy with FGF21. KEY SCIENTIFIC CONCEPTS OF REVIEW This review first illustrates the functions of FGF21, including energy metabolism, inflammation, oxidative stress, apoptosis, and autophagy. We also summarize vital functions and the underlying mechanisms through which FGF21 regulates the initiation and development of cardiac hypertrophy, connecting energy metabolism, inflammation, oxidative stress, apoptosis, and autophagy. Finally, we propose that FGF21 may be a potential therapeutic strategy for cardiac hypertrophy.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Meng Gao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), China
| | - Guohua Gong
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
6
|
Budkowska M, Ostrycharz-Jasek E, Cecerska-Heryć E, Dołęgowska K, Siennicka A, Nazarewski Ł, Rykowski P, Dołęgowska B. The Impact of Human Liver Transplantation on the Concentration of Fibroblast Growth Factors: FGF19 and FGF21. Int J Mol Sci 2025; 26:1299. [PMID: 39941067 PMCID: PMC11818808 DOI: 10.3390/ijms26031299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
The multitude of processes in which the liver participates makes it vulnerable to many serious diseases, which can lead to chronic organ failure. Modern medicine bases the treatment of end-stage liver failure on liver transplantation. To ensure the proper functioning of the transplanted liver, a balance of cellular and immunological processes and appropriate concentrations of many different factors are necessary, including, among others, fibroblast growth factors (FGFs). Over the last several years, studies have focused on some FGF growth factors, i.e., FGF19 and FGF21. These two growth factors belong to the FGF19 subfamily, and we concentrate on these two factors in our work. These factors diffuse away from the site of secretion into the blood, acting as hormones. FGF19 is a growth factor with a high therapeutic potential, involved in the homeostasis of bile acids necessary to maintain the proper function of the transplanted liver. FGF21, in turn, plays an important role in regulating lipid and glucose homeostasis. This study aimed to evaluate changes in the concentration of growth factors FGF19 and FGF21 in the plasma of 84 patients before, 24 h, and 2 weeks after liver transplantation (ELISA test was used). Additionally, the correlations of the basic laboratory parameters-alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transpeptidase (GGTP), alkaline phosphatase (ALP), total bilirubin, C-reactive protein (CRP), albumin and hemoglobin (Hb)-with FGF19 and FGF21 were determined. Our studies noted statistically significant changes in FGF19 and FGF21 concentrations before, 24 h, and 2 weeks after liver transplantation. The highest values for FGF19 before liver transplantation and the lowest values 24 h after this surgery were observed for FGF21; the highest concentrations were observed the day after liver transplantation, and the lowest were observed immediately before surgery. Observations of increases and decreases in the concentration of the examined factors at individual time points (before and after transplantation) allow us to suspect that FGF19 has an adaptive and protective function toward the transplanted liver. At the same time, FGF21 may affect the regenerative mechanisms of the damaged organ.
Collapse
Affiliation(s)
- Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Ewa Ostrycharz-Jasek
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
- Doctoral School, University of Szczecin, 70-383 Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, 71-412 Szczecin, Poland
| | - Elżbieta Cecerska-Heryć
- Department of Laboratory Medicine, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (E.C.-H.); (B.D.)
| | - Katarzyna Dołęgowska
- Department of Immunology Diagnostics, Pomeranian Medical University, Al. Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Aldona Siennicka
- Department of Medical Analytics, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Łukasz Nazarewski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, ul Banacha 1a, 02-097 Warsaw, Poland; (Ł.N.); (P.R.)
| | - Paweł Rykowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, ul Banacha 1a, 02-097 Warsaw, Poland; (Ł.N.); (P.R.)
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (E.C.-H.); (B.D.)
| |
Collapse
|
7
|
Khan MSH, Kim SQ, Ross RC, Corpodean F, Spann RA, Albarado DA, Fernandez-Kim SO, Clarke B, Berthoud HR, Münzberg H, McDougal DH, He Y, Yu S, Albaugh VL, Soto PL, Morrison CD. FGF21 acts in the brain to drive macronutrient-specific changes in behavioral motivation and brain reward signaling. Mol Metab 2025; 91:102068. [PMID: 39571902 PMCID: PMC11648240 DOI: 10.1016/j.molmet.2024.102068] [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: 09/13/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024] Open
Abstract
OBJECTIVE Dietary protein restriction induces adaptive changes in food preference, increasing protein consumption over carbohydrates or fat. We investigated whether motivation and reward signaling underpin these preferences. METHODS AND RESULTS In an operant task, protein-restricted male mice responded more for liquid protein rewards, but not carbohydrate, fat, or sweet rewards compared to non-restricted mice. When the number of responses required to access protein reward varied, protein-restricted mice exhibited higher operant responses at moderate to high response requirements. The protein restriction-induced increase in operant responding for protein was absent in Fgf21-KO mice and mice with neuron-specific deletion of the FGF21 co-receptor beta-Klotho (KlbCam2ka). Fiber photometry recording of VTA dopamine neurons revealed that oral delivery of maltodextrin triggered a larger dopamine neuron activation than casein in control diet-fed mice, while casein triggered a larger activation in low-protein diet-fed mice. This restriction-induced shift in nutrient-specific VTA dopamine signaling was lost in Fgf21-KO mice. CONCLUSION These data suggest that the increased FGF21 during protein restriction acts in the brain to induce a protein-specific appetite by specifically enhancing the reward value of protein-containing foods and the motivation to consume them.
Collapse
Affiliation(s)
| | - Sora Q Kim
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Robert C Ross
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Florina Corpodean
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Redin A Spann
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Diana A Albarado
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | | - Blaise Clarke
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | | - Heike Münzberg
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - David H McDougal
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Sangho Yu
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Vance L Albaugh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Paul L Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, 70810, USA.
| | | |
Collapse
|
8
|
de La Bourdonnaye G, Ghazalova T, Fojtik P, Kutalkova K, Bednar D, Damborsky J, Rotrekl V, Stepankova V, Chaloupkova R. Computer-aided engineering of stabilized fibroblast growth factor 21. Comput Struct Biotechnol J 2024; 23:942-951. [PMID: 38379823 PMCID: PMC10877085 DOI: 10.1016/j.csbj.2024.02.001] [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: 10/09/2023] [Revised: 02/03/2024] [Accepted: 02/03/2024] [Indexed: 02/22/2024] Open
Abstract
FGF21 is an endocrine signaling protein belonging to the family of fibroblast growth factors (FGFs). It has emerged as a molecule of interest for treating various metabolic diseases due to its role in regulating glucogenesis and ketogenesis in the liver. However, FGF21 is prone to heat, proteolytic, and acid-mediated degradation, and its low molecular weight makes it susceptible to kidney clearance, significantly reducing its therapeutic potential. Protein engineering studies addressing these challenges have generally shown that increasing the thermostability of FGF21 led to improved pharmacokinetics. Here, we describe the computer-aided design and experimental characterization of FGF21 variants with enhanced melting temperature up to 15 °C, uncompromised efficacy at activation of MAPK/ERK signaling in Hep G2 cell culture, and ability to stimulate proliferation of Hep G2 and NIH 3T3 fibroblasts cells comparable with FGF21-WT. We propose that stabilizing the FGF21 molecule by rational design should be combined with other reported stabilization strategies to maximize the pharmaceutical potential of FGF21.
Collapse
Affiliation(s)
- Gabin de La Bourdonnaye
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Enantis Ltd., Biotechnology Incubator INBIT, Brno, Czech Republic
| | - Tereza Ghazalova
- Enantis Ltd., Biotechnology Incubator INBIT, Brno, Czech Republic
| | - Petr Fojtik
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | - David Bednar
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Loschmidt Laboratories, Centre for Toxic Compounds in the Environment RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jiri Damborsky
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Loschmidt Laboratories, Centre for Toxic Compounds in the Environment RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Vladimir Rotrekl
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | | | - Radka Chaloupkova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Enantis Ltd., Biotechnology Incubator INBIT, Brno, Czech Republic
| |
Collapse
|
9
|
Meroni M, Dongiovanni P, Tiano F, Piciotti R, Alisi A, Panera N. β-Klotho as novel therapeutic target in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A narrative review. Biomed Pharmacother 2024; 180:117608. [PMID: 39490050 DOI: 10.1016/j.biopha.2024.117608] [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: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) represents the most frequent cause of hepatic disorder, and its progressive form defined as Metabolic Dysfunction-Associated Steatohepatitis (MASH) contributes to the development of fibrosis/cirrhosis and hepatocellular carcinoma (HCC). Today effective therapeutic strategies addressing MASH-related comorbidities, inflammation, and fibrosis are needed. The fibroblast growth factor (FGF) 19 and 21 and their fibroblast growth factor receptor/β-Klotho (KLB) complexes have recently emerged as promising druggable targets for MASLD. However, less is known regarding the causative association between KLB activity and advanced stages of liver disease. In the present narrative review, we aimed to provide an up-to-date picture of the role of the KLB co-receptor in MASLD development and progression. We performed a detailed analysis of recently published preclinical and clinical data to decipher the molecular mechanisms underlying KLB function and to correlate the presence of inherited or acquired KLB aberrancies with the predisposition towards MASLD. Moreover, we described ongoing clinical trials evaluating the therapeutic approaches targeting FGF19-21/FGFR/KLB in patients with MASLD and discussed the challenges related to their use. We furtherly described that KLB exhibits protective effects against metabolic disorders by acting in an FGF-dependent and independent manner thus triggering the hypothesis that KLB soluble forms may play a critical role in preserving liver health. Therefore, targeting KLB may provide promising strategies for treating MASLD, as supported by experimental evidence and ongoing clinical trials.
Collapse
Affiliation(s)
- Marica Meroni
- Medicine and Metabolic Diseases; Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases; Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Francesca Tiano
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Roberto Piciotti
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan 20122, Italy
| | - Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Nadia Panera
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
10
|
Morita H, Hoshiga M. Fibroblast Growth Factors in Cardiovascular Disease. J Atheroscler Thromb 2024; 31:1496-1511. [PMID: 39168622 PMCID: PMC11537794 DOI: 10.5551/jat.rv22025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Despite advancements in managing traditional cardiovascular risk factors, many cardiovascular diseases (CVDs) persist. Fibroblast growth factors (FGFs) have emerged as potential diagnostic markers and therapeutic targets for CVDs. FGF1, FGF2, and FGF4 are primarily used for therapeutic angiogenesis. Clinical applications are being explored based on animal studies using approaches such as recombinant protein administration and adenovirus-mediated gene delivery, targeting patients with coronary artery disease and lower extremity arterial disease. Although promising results have been observed in animal models and early-stage clinical trials, further studies are required to assess their therapeutic potential. The FGF19 subfamily, consisting of FGF19, FGF21, and FGF23, act via endocrine signaling in various organs. FGF19, primarily expressed in the small intestine, plays important roles in glucose, lipid, and bile acid metabolism and has therapeutic potential for metabolic disorders. FGF21, found in various tissues, improves glucose metabolism and insulin sensitivity, suggesting potential for treating obesity and diabetes. FGF23, primarily secreted by osteocytes, regulates vitamin D and phosphate metabolism and serves as an important biomarker for chronic kidney disease and CVDs. Thus, FGFs holds promise for both therapeutic and diagnostic applications in metabolic and cardiovascular diseases. Understanding the mechanisms of FGF may pave the way for novel strategies to prevent and manage CVDs, potentially addressing the limitations of current treatments. This review explores the roles of FGF1, FGF2, FGF4, and the FGF19 subfamily in maintaining cardiovascular health. Further research and clinical trials are crucial to fully understand the therapeutic potential of FGFs in managing cardiovascular health.
Collapse
Affiliation(s)
- Hideaki Morita
- Department of Cardiology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Masaaki Hoshiga
- Department of Cardiology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
11
|
Kanbay M, Mutlu A, Bakir CN, Peltek IB, Canbaz AA, Díaz Tocados JM, Haarhaus M. Klotho in pregnancy and intrauterine development-potential clinical implications: a review from the European Renal Association CKD-MBD Working Group. Nephrol Dial Transplant 2024; 39:1574-1582. [PMID: 38486352 PMCID: PMC11427066 DOI: 10.1093/ndt/gfae066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Indexed: 09/28/2024] Open
Abstract
Intrauterine development is crucial for life-long health; therefore, elucidation of its key regulators is of interest for their potential prognostic and therapeutic implications. Originally described as a membrane-bound anti-aging protein, Klotho has evolved as a regulator of numerous functions in different organ systems. Circulating Klotho is generated by alternative splicing or active shedding from cell membranes. Recently, Klotho was identified as a regulator of placental function, and while Klotho does not cross the placental barrier, increased levels of circulating α-Klotho have been identified in umbilical cord blood compared with maternal blood, indicating that Klotho may also play a role in intrauterine development. In this narrative review, we discuss novel insights into the specific functions of the Klotho proteins in the placenta and in intrauterine development, while summarizing up-to-date knowledge about their structures and functions. Klotho plays a role in stem cell functioning, organogenesis and haematopoiesis. Low circulating maternal and foetal levels of Klotho are associated with preeclampsia, intrauterine growth restriction, and an increased perinatal risk for newborns, indicating a potential use of Klotho as biomarker and therapeutic target. Experimental administration of Klotho protein indicates a neuro- and nephroprotective potential, suggesting a possible future role of Klotho as a therapeutic agent. However, the use of Klotho as intervention during pregnancy is as yet unproven. Here, we summarize novel evidence, suggesting Klotho as a key regulator for healthy pregnancies and intrauterine development with promising potential for clinical use.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - Ali Mutlu
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Cicek N Bakir
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ibrahim B Peltek
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ata A Canbaz
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Juan Miguel Díaz Tocados
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida, Dr Pifarré Foundation (IRBLleida), Lleida, Spain
| | - Mathias Haarhaus
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
- Diaverum AB, Malmö, Sweden
| |
Collapse
|
12
|
Sullivan AI, Jensen-Cody SO, Claflin KE, Vorhies KE, Flippo KH, Potthoff MJ. Characterization of FGF21 Sites of Production and Signaling in Mice. Endocrinology 2024; 165:bqae120. [PMID: 39253796 DOI: 10.1210/endocr/bqae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/08/2024] [Accepted: 09/08/2024] [Indexed: 09/11/2024]
Abstract
Fibroblast growth factor (FGF) 21 is an endocrine hormone that signals to multiple tissues to regulate metabolism. FGF21 and another endocrine FGF, FGF15/19, signal to target tissues by binding to the co-receptor β-klotho (KLB), which then facilitates the interaction of these different FGFs with their preferred FGF receptor. KLB is expressed in multiple metabolic tissues, but the specific cell types and spatial distribution of these cells are not known. Furthermore, while circulating FGF21 is primarily produced by the liver, recent publications have indicated that brain-derived FGF21 impacts memory and learning. Here we use reporter mice to comprehensively assess KLB and FGF21 expression throughout the body. These data provide an important resource for guiding future studies to identify important peripheral and central targets of FGFs and to determine the significance of nonhepatic FGF21 production.
Collapse
Affiliation(s)
- Andrew I Sullivan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Sharon O Jensen-Cody
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kristin E Claflin
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kai E Vorhies
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kyle H Flippo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
| |
Collapse
|
13
|
Zhang T, Jiang D, Zhang X, Chen L, Jiang J, Zhang C, Li S, Li Q. The role of nonmyocardial cells in the development of diabetic cardiomyopathy and the protective effects of FGF21: a current understanding. Cell Commun Signal 2024; 22:446. [PMID: 39327594 PMCID: PMC11426003 DOI: 10.1186/s12964-024-01842-0] [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: 06/05/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) represents a unique myocardial disease originating from diabetic metabolic disturbances that is characterized by myocardial fibrosis and diastolic dysfunction. While recent research regarding the pathogenesis and treatment of DCM has focused primarily on myocardial cells, nonmyocardial cells-including fibroblasts, vascular smooth muscle cells (VSMCs), endothelial cells (ECs), and immune cells-also contribute significantly to the pathogenesis of DCM. Among various therapeutic targets, fibroblast growth factor 21 (FGF21) has been identified as a promising agent because of its cardioprotective effects that extend to nonmyocardial cells. In this review, we aim to elucidate the role of nonmyocardial cells in DCM and underscore the potential of FGF21 as a therapeutic strategy for these cells.
Collapse
Affiliation(s)
- Tianyi Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Donghui Jiang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiao Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Shengbiao Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Qiuhong Li
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
14
|
Kim SQ, Spann RA, Khan MSH, Berthoud HR, Münzberg H, Albaugh VL, He Y, McDougal DH, Soto P, Yu S, Morrison CD. FGF21 as a mediator of adaptive changes in food intake and macronutrient preference in response to protein restriction. Neuropharmacology 2024; 255:110010. [PMID: 38797244 PMCID: PMC11156534 DOI: 10.1016/j.neuropharm.2024.110010] [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: 03/20/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Free-feeding animals navigate complex nutritional landscapes in which food availability, cost, and nutritional value can vary markedly. Animals have thus developed neural mechanisms that enable the detection of nutrient restriction, and these mechanisms engage adaptive physiological and behavioral responses that limit or reverse this nutrient restriction. This review focuses specifically on dietary protein as an essential and independently defended nutrient. Adequate protein intake is required for life, and ample evidence exists to support an active defense of protein that involves behavioral changes in food intake, food preference, and food motivation, likely mediated by neural changes that increase the reward value of protein foods. Available evidence also suggests that the circulating hormone fibroblast growth factor 21 (FGF21) acts in the brain to coordinate these adaptive changes in food intake, making it a unique endocrine signal that drives changes in macronutrient preference in the context of protein restriction. This article is part of the Special Issue on "Food intake and feeding states".
Collapse
Affiliation(s)
- Sora Q Kim
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Redin A Spann
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | | | | - Heike Münzberg
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Vance L Albaugh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - David H McDougal
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Paul Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, 70810, USA
| | - Sangho Yu
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | |
Collapse
|
15
|
Trusz GJ. Fibroblast growth factor 21. Differentiation 2024; 139:100793. [PMID: 38991938 DOI: 10.1016/j.diff.2024.100793] [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: 09/01/2023] [Revised: 06/23/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
Fibroblast growth factor 21 (FGF21) belongs to the FGF19 subfamily and acts systemically, playing a key role in inter-organ crosstalk. Ranging from metabolism, reproduction, and immunity, FGF21 is a pleiotropic hormone which contributes to various physiological processes. Although most of its production across species stems from hepatic tissues, expression of FGF21 in mice has also been identified in adipose tissue, thymus, heart, pancreas, and skeletal muscle. Elevated FGF21 levels are affiliated with various diseases and conditions, such as obesity, type 2 diabetes, preeclampsia, as well as cancer. Murine knockout models are viable and show modest weight gain, while overexpression and gain-of-function models display resistance to weight gain, altered bone volume, and enhanced immunity. In addition, FGF21-based therapies are at the forefront of biopharmaceutical strategies aimed at treating metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Guillaume J Trusz
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| |
Collapse
|
16
|
Khan MSH, Kim SQ, Ross RC, Corpodean F, Spann RA, Albarado DA, Fernandez-Kim SO, Clarke B, Berthoud HR, Münzberg H, McDougal DH, He Y, Yu S, Albaugh VL, Soto P, Morrison CD. FGF21 acts in the brain to drive macronutrient-specific changes in behavioral motivation and brain reward signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583399. [PMID: 38798313 PMCID: PMC11118293 DOI: 10.1101/2024.03.05.583399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Dietary protein restriction induces adaptive changes in food preference, increasing protein consumption over carbohydrates or fat. We investigated whether motivation and reward signaling underpin these preferences. In an operant task, protein-restricted male mice responded more for liquid protein rewards, but not carbohydrate, fat, or sweet rewards compared to non-restricted mice. The protein restriction-induced increase in operant responding for protein was absent in Fgf21-KO mice and mice with neuron-specific deletion of the FGF21 co-receptor beta-Klotho (Klb Cam2ka ) mice. Fiber photometry recording of VTA dopamine neurons revealed that oral delivery of maltodextrin triggered a larger activation as compared to casein in control-fed mice, whereas casein triggered a larger activation in protein-restricted mice. This restriction-induced shift in nutrient-specific VTA dopamine signaling was lost in Fgf21-KO mice. These data strongly suggest that the increased FGF21 during protein restriction acts in the brain to induce a protein-specific appetite by specifically enhancing the reward value of protein-containing foods and the motivation to consume them.
Collapse
Affiliation(s)
| | - Sora Q. Kim
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Robert C. Ross
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Florina Corpodean
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Redin A. Spann
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | | | | | - Blaise Clarke
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | | | - Heike Münzberg
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | | | - Yanlin He
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Sangho Yu
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Vance L. Albaugh
- Pennington Biomedical Research Center, Baton Rouge, LA 70808
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Paul Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA 70810
| | | |
Collapse
|
17
|
Phan P, Ternier G, Edirisinghe O, Kumar TKS. Exploring endocrine FGFs - structures, functions and biomedical applications. INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 15:68-99. [PMID: 39309613 PMCID: PMC11411148 DOI: 10.62347/palk2137] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024]
Abstract
The family of fibroblast growth factors (FGFs) consists of 22 members with diverse biological functions in cells, from cellular development to metabolism. The family can be further categorized into three subgroups based on their three modes of action. FGF19, FGF21, and FGF23 are endocrine FGFs that act in a hormone-like/endocrine manner to regulate various metabolic activities. However, all three members of the endocrine family require both FGF receptors (FGFRs) and klotho co-receptors to elicit their functions. α-klotho and β-klotho act as scaffolds to bring endocrine FGFs closer to their receptors (FGFRs) to form active complexes. Numerous novel studies about metabolic FGFs' structures, mechanisms, and physiological insights have been published to further understand the complex molecular interactions and physiological activities of endocrine FGFs. Herein, we aim to review the structures, physiological functions, binding mechanisms to cognate receptors, and novel biomedical applications of endocrine FGFs in recent years.
Collapse
Affiliation(s)
- Phuc Phan
- Department of Chemistry and Biochemistry, Fulbright College of Art and Sciences, University of ArkansasFayetteville, AR 72701, USA
| | - Gaёtane Ternier
- Department of Chemistry and Biochemistry, Fulbright College of Art and Sciences, University of ArkansasFayetteville, AR 72701, USA
| | - Oshadi Edirisinghe
- Cell and Molecular Biology Program, University of ArkansasFayetteville, AR 72701, USA
| | | |
Collapse
|
18
|
Habib S. Team players in the pathogenesis of metabolic dysfunctions-associated steatotic liver disease: The basis of development of pharmacotherapy. World J Gastrointest Pathophysiol 2024; 15:93606. [PMID: 39220834 PMCID: PMC11362842 DOI: 10.4291/wjgp.v15.i4.93606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
Nutrient metabolism is regulated by several factors. Social determinants of health with or without genetics are the primary regulator of metabolism, and an unhealthy lifestyle affects all modulators and mediators, leading to the adaptation and finally to the exhaustion of cellular functions. Hepatic steatosis is defined by presence of fat in more than 5% of hepatocytes. In hepatocytes, fat is stored as triglycerides in lipid droplet. Hepatic steatosis results from a combination of multiple intracellular processes. In a healthy individual nutrient metabolism is regulated at several steps. It ranges from the selection of nutrients in a grocery store to the last step of consumption of ATP as an energy or as a building block of a cell as structural component. Several hormones, peptides, and genes have been described that participate in nutrient metabolism. Several enzymes participate in each nutrient metabolism as described above from ingestion to generation of ATP. As of now several publications have revealed very intricate regulation of nutrient metabolism, where most of the regulatory factors are tied to each other bidirectionally, making it difficult to comprehend chronological sequence of events. Insulin hormone is the primary regulator of all nutrients' metabolism both in prandial and fasting states. Insulin exerts its effects directly and indirectly on enzymes involved in the three main cellular function processes; metabolic, inflammation and repair, and cell growth and regeneration. Final regulators that control the enzymatic functions through stimulation or suppression of a cell are nuclear receptors in especially farnesoid X receptor and peroxisome proliferator-activated receptor/RXR ligands, adiponectin, leptin, and adiponutrin. Insulin hormone has direct effect on these final modulators. Whereas blood glucose level, serum lipids, incretin hormones, bile acids in conjunction with microbiota are intermediary modulators which are controlled by lifestyle. The purpose of this review is to overview the key players in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) that help us understand the disease natural course, risk stratification, role of lifestyle and pharmacotherapy in each individual patient with MASLD to achieve personalized care and target the practice of precision medicine. PubMed and Google Scholar databases were used to identify publication related to metabolism of carbohydrate and fat in states of health and disease states; MASLD, cardiovascular disease and cancer. More than 1000 publications including original research and review papers were reviewed.
Collapse
Affiliation(s)
- Shahid Habib
- Department of Hepatology, Liver Institute PLLC, Tucson, AZ 85712, United States
| |
Collapse
|
19
|
Bartmańska M, Wiecek A, Adamczak M. Plasma FGF21 Concentration in Kidney Transplant Patients-Results from Prospective and Cross-Sectional Studies. J Clin Med 2024; 13:4266. [PMID: 39064306 PMCID: PMC11278288 DOI: 10.3390/jcm13144266] [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: 05/28/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Background/Objectives: Fibroblast growth factor 21 (FGF21) is a protein hormone involved in physiological conditions in the regulation of energy expenditure and several metabolic processes. The aim of this present study was to analyze the effect of successful kidney transplantations on the plasma FGF21 concentration and to study the factors which may influence plasma FGF21 concentration in patients in long time after kidney transplantation. Methods: This study consisted of two independent parts. The first part was a prospective observation of CKD patients in stage 5 before and then on the 14th and 30th day and 6 months after kidney transplantation. The second part of this study was the cross-sectional study completed in patients at least one year after kidney transplantation and the control group. In CKD patients directly before and during the early period after KTx, plasma FGF21 concentrations were measured four times (immediately before and 14 and 30 days and 6 months after KTx). In patients long time after kidney transplantation and in healthy subjects, plasma FGF21 concentration was measured once. Results: Forty patients with chronic kidney disease (CKD) who were either directly before or within the early period after kidney transplantation (KTx), 184 patients longtime after KTx and 50 healthy subjects were enrolled into this study. In CKD patients at the stage directly before receiving a KTx, the mean plasma FGF21 concentration was significantly higher than in the healthy subjects [1013.0 pg/mL versus 239.5 pg/mL, p < 0.001]. At 14, 30 days, and 6 months after the KTx, a significant decrease of plasma FGF21 was observed, with values of 322.5 pg/mL; 355.0 pg/mL; and 344.0 pg/mL (p < 0.001), respectively]. In patients long time after KTx, a negative correlation was found between the plasma FGF21 concentration and the estimated glomerular filtration rate and a positive correlation was found between the plasma FGF21 concentration and the BMI, the serum concentration of triglycerides, insulin, interleukin-6, CRP, and cystatin C. Conclusions: The plasma FGF21 concentration in patients with end-stage renal disease is higher than in healthy subjects and significantly decreases after a successful KTx. The plasma FGF21 concentration measured by ELISA in patients long time after kidney transplantation seems to be related to the degree of kidney function impairment and their metabolic status. The kidneys appear to be one of the main organs involved in the biodegradation and/or elimination of FGF21.
Collapse
Affiliation(s)
| | | | - Marcin Adamczak
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland (A.W.)
| |
Collapse
|
20
|
Ma Y, Singhal G, Chan SS, Wang C, Yu H, Yin B, Pang J, Malvar G, Nasser I, Mather ML, Maratos-Flier E. FGF21 protects against ischaemia reperfusion injury in normal and fatty livers. Liver Int 2024; 44:1668-1679. [PMID: 38554044 DOI: 10.1111/liv.15911] [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: 08/08/2023] [Revised: 03/02/2024] [Accepted: 03/11/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND Liver ischaemia/reperfusion (I/R) injury, which is an inevitable clinical problem of liver resection, liver transplantation and haemorrhagic shock. Fibroblast growth factor 21 (FGF21) was intimately coupled with multiple metabolic processes and proved to protect against apoptosis and inflammatory response in hepatocytes during hepatic I/R injury. However, the regulatory mechanisms of FGF21 in hepatic I/R injury remains unknown. Therefore, we hypothesize that FGF21 protects hepatic tissues from I/R injury. METHODS Blood samples were available from haemangiomas patients undergoing hepatectomy and murine liver I/R model and used to further evaluate the serum levels of FGF21 both in humans and mice. We further explored the regulatory mechanisms of FGF21 in murine liver I/R model by using FGF21-knockout mice (FGF21-KO mice) and FGF21-overexpression transgenic mice (FGF21-OE mice) fed a high-fat or ketogenic diet. RESULTS Our results show that the circulating levels of FGF21 were robustly decreased after liver I/R in both humans and mice. Silencing FGF21 expression with FGF21-KO mice aggravates liver injury at 6 h after 75 min of partial liver ischaemia, while FGF21-OE mice display alleviated hepatic I/R injury and inflammatory response. Compared with chow diet mice, exogenous FGF21 decreases the levels of aminotransferase, histological changes, apoptosis and inflammatory response in hepatic I/R injury treatment mice with a high-fat diet. Meanwhile, ketogenic diet mice are not sensitive to hepatic I/R injury. CONCLUSIONS The circulating contents of FGF21 are decreased during liver warm I/R injury and exogenous FGF21 exerts hepatoprotective effects on hepatic I/R injury. Thus, FGF21 regulates hepatic I/R injury and may be a key therapeutic target.
Collapse
Affiliation(s)
- Yong Ma
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Garima Singhal
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Suzanne S Chan
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Chaoqun Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongjun Yu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bing Yin
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of Minimal Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jing Pang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Grace Malvar
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Imad Nasser
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Marie L Mather
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Eleftheria Maratos-Flier
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Negroiu CE, Tudoraşcu RI, Beznă MC, Ungureanu AI, Honţaru SO, Dănoiu S. The role of FGF21 in the interplay between obesity and non-alcoholic fatty liver disease: a narrative review. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:159-172. [PMID: 39020530 PMCID: PMC11384831 DOI: 10.47162/rjme.65.2.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
Obesity poses a significant and escalating challenge in contemporary society, increasing the risk of developing various metabolic disorders such as dyslipidemia, cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), type 2 diabetes, and certain types of cancer. The current array of therapeutic interventions for obesity remains insufficient, prompting a pressing demand for novel and more effective treatments. In response, scientific attention has turned to the fibroblast growth factor 21 (FGF21) due to its remarkable and diverse impacts on lipid, carbohydrate, and energy metabolism. This comprehensive review aims to delve into the multifaceted aspects of FGF21, encompassing its discovery, synthesis, functional roles, and potential as a biomarker and therapeutic agent, with a specific focus on its implications for NAFLD.
Collapse
Affiliation(s)
- Cristina Elena Negroiu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, Romania; ; Department of Health Care and Physiotherapy, Faculty of Sciences, Physical Education and Informatics, University Center of Piteşti, National University for Science and Technology Politehnica, Bucharest, Romania;
| | | | | | | | | | | |
Collapse
|
22
|
Hu C, Qiao W, Li X, Ning ZK, Liu J, Dalangood S, Li H, Yu X, Zong Z, Wen Z, Gui J. Tumor-secreted FGF21 acts as an immune suppressor by rewiring cholesterol metabolism of CD8 +T cells. Cell Metab 2024; 36:630-647.e8. [PMID: 38309268 DOI: 10.1016/j.cmet.2024.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/19/2023] [Accepted: 01/10/2024] [Indexed: 02/05/2024]
Abstract
Tumors employ diverse strategies for immune evasion. Unraveling the mechanisms by which tumors suppress anti-tumor immunity facilitates the development of immunotherapies. Here, we have identified tumor-secreted fibroblast growth factor 21 (FGF21) as a pivotal immune suppressor. FGF21 is upregulated in multiple types of tumors and promotes tumor progression. Tumor-secreted FGF21 significantly disrupts anti-tumor immunity by rewiring cholesterol metabolism of CD8+T cells. Mechanistically, FGF21 sustains the hyperactivation of AKT-mTORC1-sterol regulatory-element-binding protein 1 (SREBP1) signal axis in the activated CD8+T cells, resulting in the augment of cholesterol biosynthesis and T cell exhaustion. FGF21 knockdown or blockade using a neutralizing antibody normalizes AKT-mTORC1 signaling and reduces excessive cholesterol accumulation in CD8+T cells, thus restoring CD8+T cytotoxic function and robustly suppressing tumor growth. Our findings reveal FGF21 as a "secreted immune checkpoint" that hampers anti-tumor immunity, suggesting that inhibiting FGF21 could be a valuable strategy to enhance the cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Cegui Hu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wen Qiao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiang Li
- Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhi-Kun Ning
- Department of Gastroenterological Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jiang Liu
- Department of Gastroenterological Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Sumiya Dalangood
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hanjun Li
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiang Yu
- Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhen Zong
- Department of Gastroenterological Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China.
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China.
| | - Jun Gui
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
23
|
Richter MM, Thomsen MN, Skytte MJ, Kjeldsen SAS, Samkani A, Frystyk J, Magkos F, Holst JJ, Madsbad S, Krarup T, Haugaard SB, Wewer Albrechtsen NJ. Effect of a 6-Week Carbohydrate-Reduced High-Protein Diet on Levels of FGF21 and GDF15 in People With Type 2 Diabetes. J Endocr Soc 2024; 8:bvae008. [PMID: 38379856 PMCID: PMC10875725 DOI: 10.1210/jendso/bvae008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Indexed: 02/22/2024] Open
Abstract
Context Fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) are increased in type 2 diabetes and are potential regulators of metabolism. The effect of changes in caloric intake and macronutrient composition on their circulating levels in patients with type 2 diabetes are unknown. Objective To explore the effects of a carbohydrate-reduced high-protein diet with and without a clinically significant weight loss on circulating levels of FGF21 and GDF15 in patients with type 2 diabetes. Methods We measured circulating FGF21 and GDF15 in patients with type 2 diabetes who completed 2 previously published diet interventions. Study 1 randomized 28 subjects to an isocaloric diet in a 6 + 6-week crossover trial consisting of, in random order, a carbohydrate-reduced high-protein (CRHP) or a conventional diabetes (CD) diet. Study 2 randomized 72 subjects to a 6-week hypocaloric diet aiming at a ∼6% weight loss induced by either a CRHP or a CD diet. Fasting plasma FGF21 and GDF15 were measured before and after the interventions in a subset of samples (n = 24 in study 1, n = 66 in study 2). Results Plasma levels of FGF21 were reduced by 54% in the isocaloric study (P < .05) and 18% in the hypocaloric study (P < .05) in CRHP-treated individuals only. Circulating GDF15 levels increased by 18% (P < .05) following weight loss in combination with a CRHP diet but only in those treated with metformin. Conclusion The CRHP diet significantly reduced FGF21 in people with type 2 diabetes independent of weight loss, supporting the role of FGF21 as a "nutrient sensor." Combining metformin treatment with carbohydrate restriction and weight loss may provide additional metabolic improvements due to the rise in circulating GDF15.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Mads N Thomsen
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
| | - Mads J Skytte
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Department of Forensic Medicine, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Amirsalar Samkani
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
| | - Jan Frystyk
- Department of Endocrinology, Odense University Hospital, Odense, 5000, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital—Hvidovre, Hvidovre, 2650, Denmark
| | - Thure Krarup
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg, Copenhagen, 2400, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| |
Collapse
|
24
|
Yang Z, Zarbl H, Guo GL. Circadian Regulation of Endocrine Fibroblast Growth Factors on Systemic Energy Metabolism. Mol Pharmacol 2024; 105:179-193. [PMID: 38238100 PMCID: PMC10877735 DOI: 10.1124/molpharm.123.000831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/05/2024] [Indexed: 02/17/2024] Open
Abstract
The circadian clock is an endogenous biochemical timing system that coordinates the physiology and behavior of organisms to earth's ∼24-hour circadian day/night cycle. The central circadian clock synchronized by environmental cues hierarchically entrains peripheral clocks throughout the body. The circadian system modulates a wide variety of metabolic signaling pathways to maintain whole-body metabolic homeostasis in mammals under changing environmental conditions. Endocrine fibroblast growth factors (FGFs), namely FGF15/19, FGF21, and FGF23, play an important role in regulating systemic metabolism of bile acids, lipids, glucose, proteins, and minerals. Recent evidence indicates that endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between peripheral clocks and energy homeostasis by regulating the expression of metabolic enzymes and hormones. Circadian disruption induced by environmental stressors or genetic ablation is associated with metabolic dysfunction and diurnal disturbances in FGF signaling pathways that contribute to the pathogenesis of metabolic diseases. Time-restricted feeding strengthens the circadian pattern of metabolic signals to improve metabolic health and prevent against metabolic diseases. Chronotherapy, the strategic timing of medication administration to maximize beneficial effects and minimize toxic effects, can provide novel insights into linking biologic rhythms to drug metabolism and toxicity within the therapeutical regimens of diseases. Here we review the circadian regulation of endocrine FGF signaling in whole-body metabolism and the potential effect of circadian dysfunction on the pathogenesis and development of metabolic diseases. We also discuss the potential of chrononutrition and chronotherapy for informing the development of timing interventions with endocrine FGFs to optimize whole-body metabolism in humans. SIGNIFICANCE STATEMENT: The circadian timing system governs physiological, metabolic, and behavioral functions in living organisms. The endocrine fibroblast growth factor (FGF) family (FGF15/19, FGF21, and FGF23) plays an important role in regulating energy and mineral metabolism. Endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between circadian clocks and metabolic homeostasis. Chronic disruption of circadian rhythms increases the risk of metabolic diseases. Chronological interventions such as chrononutrition and chronotherapy provide insights into linking biological rhythms to disease prevention and treatment.
Collapse
Affiliation(s)
- Zhenning Yang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (Z.Y., G.L.G.), Environmental and Occupational Health Sciences Institute (Z.Y., H.Z., G.L.G.), Department of Environmental and Occupational Health Justice, School of Public Health (H.Z.), Rutgers Center for Lipid Research (G.L.G.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.)
| | - Helmut Zarbl
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (Z.Y., G.L.G.), Environmental and Occupational Health Sciences Institute (Z.Y., H.Z., G.L.G.), Department of Environmental and Occupational Health Justice, School of Public Health (H.Z.), Rutgers Center for Lipid Research (G.L.G.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.)
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (Z.Y., G.L.G.), Environmental and Occupational Health Sciences Institute (Z.Y., H.Z., G.L.G.), Department of Environmental and Occupational Health Justice, School of Public Health (H.Z.), Rutgers Center for Lipid Research (G.L.G.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.)
| |
Collapse
|
25
|
Zangerolamo L, Carvalho M, Velloso LA, Barbosa HCL. Endocrine FGFs and their signaling in the brain: Relevance for energy homeostasis. Eur J Pharmacol 2024; 963:176248. [PMID: 38056616 DOI: 10.1016/j.ejphar.2023.176248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Since their discovery in 2000, there has been a continuous expansion of studies investigating the physiology, biochemistry, and pharmacology of endocrine fibroblast growth factors (FGFs). FGF19, FGF21, and FGF23 comprise a subfamily with attributes that distinguish them from typical FGFs, as they can act as hormones and are, therefore, referred to as endocrine FGFs. As they participate in a broad cross-organ endocrine signaling axis, endocrine FGFs are crucial lipidic, glycemic, and energetic metabolism regulators during energy availability fluctuations. They function as powerful metabolic signals in physiological responses induced by metabolic diseases, like type 2 diabetes and obesity. Pharmacologically, FGF19 and FGF21 cause body weight loss and ameliorate glucose homeostasis and energy expenditure in rodents and humans. In contrast, FGF23 expression in mice and humans has been linked with insulin resistance and obesity. Here, we discuss emerging concepts in endocrine FGF signaling in the brain and critically assess their putative role as therapeutic targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Lucas Zangerolamo
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Marina Carvalho
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Helena C L Barbosa
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil.
| |
Collapse
|
26
|
Wu M, Hu C, Sun T, Xun L, Zhao Q. Effective fraction of Gualou-Xiebai-Banxia decoction inhibits the apoptosis of myocardial cells induced by ox-LDL via FGF21/FGFR1/βKlotho-FRS2α signal pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117054. [PMID: 37595815 DOI: 10.1016/j.jep.2023.117054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Gualou-Xiebai-Banxia decoction (GXBD), a classical traditional Chinese medicine (TCM) formula, has beneficial effects in turbid phlegm obstruction syndrome, a type of coronary heart disease (CHD). However, the underlying mechanism and effective constituents of GXBD remain elusive. Our previous studies have shown that the effective constituents of GXBD may be enriched in the n-butanol fraction (GXB-N) and water fraction (GXB-W), the targets of which remain unknown. AIM OF THE STUDY To investigate whether GXB-N and GXB-W protect myocardial cells (MCs) via fibroblast growth factor 21 (FGF21) signaling and, if so, to elucidate the underlying mechanisms. Furthermore, to investigate the targets of GXB-N and GXB-W as potential therapeutic targets for cardiovascular disease (CVD). MATERIALS AND METHODS Cell viability and apoptosis were assayed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays, respectively. The content of FGF21 in the medium was measured using enzyme-linked immunosorbent assay (ELISA). Protein expression was detected using immunofluorescence and western blotting. RESULTS Apoptosis increased markedly in MCs exposed to oxidized low density lipoprotein (ox-LDL) 100 μg/mL, with increased expression of FGF21, FGFR1 and βKlotho, phosphorylation of fibroblast receptor substrate 2α (FRS2α) was suppressed. Following incubation with GXB-N and GXB-W 200 μg/mL, the expression of FGF21, FGFR1, and βKlotho and the phosphorylation of FRS2α were increased. CONCLUSION Ox-LDL may inhibit the phosphorylation of FRS2α, inducing considerable FGF21 resistance and resulting in MC apoptosis. GXB-N and GXB-W restored and enhanced FGF21 sensitivity in MCs, consequently rescuing cells from ox-LDL-induced apoptosis. The FGF21-FRS2α signal pathway may be part action targets of these two effective fractions of GXBD.
Collapse
Affiliation(s)
- Mengxue Wu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Chaoqun Hu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Teng Sun
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Liying Xun
- School of Basic Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Qitao Zhao
- School of Basic Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
27
|
Sadowska A, Poniedziałek-Czajkowska E, Mierzyński R. The Role of the FGF19 Family in the Pathogenesis of Gestational Diabetes: A Narrative Review. Int J Mol Sci 2023; 24:17298. [PMID: 38139126 PMCID: PMC10743406 DOI: 10.3390/ijms242417298] [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: 10/27/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common pregnancy complications. Understanding the pathogenesis and appropriate diagnosis of GDM enables the implementation of early interventions during pregnancy that reduce the risk of maternal and fetal complications. At the same time, it provides opportunities to prevent diabetes, metabolic syndrome, and cardiovascular diseases in women with GDM and their offspring in the future. Fibroblast growth factors (FGFs) represent a heterogeneous family of signaling proteins which play a vital role in cell proliferation and differentiation, repair of damaged tissues, wound healing, angiogenesis, and mitogenesis and also affect the regulation of carbohydrate, lipid, and hormone metabolism. Abnormalities in the signaling function of FGFs may lead to numerous pathological conditions, including metabolic diseases. The FGF19 subfamily, also known as atypical FGFs, which includes FGF19, FGF21, and FGF23, is essential in regulating metabolic homeostasis and acts as a hormone while entering the systemic circulation. Many studies have pointed to the involvement of the FGF19 subfamily in the pathogenesis of metabolic diseases, including GDM, although the results are inconclusive. FGF19 and FGF21 are thought to be associated with insulin resistance, an essential element in the pathogenesis of GDM. FGF21 may influence placental metabolism and thus contribute to fetal growth and metabolism regulation. The observed relationship between FGF21 and increased birth weight could suggest a potential role for FGF21 in predicting future metabolic abnormalities in children born to women with GDM. In this group of patients, different mechanisms may contribute to an increased risk of cardiovascular diseases in women in later life, and FGF23 appears to be their promising early predictor. This study aims to present a comprehensive review of the FGF19 subfamily, emphasizing its role in GDM and predicting its long-term metabolic consequences for mothers and their offspring.
Collapse
Affiliation(s)
| | - Elżbieta Poniedziałek-Czajkowska
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland; (A.S.); (R.M.)
| | | |
Collapse
|
28
|
Liu Z, Peng Y, Li S, Lin Y, Huang Y, Chen W, Bao C, Zhou Z, Lin Z, Chen L. Increased circulating FGF21 level predicts the burden of metabolic demands and risk of vascular diseases in adults with type 2 diabetes. BMC Endocr Disord 2023; 23:272. [PMID: 38057786 PMCID: PMC10702049 DOI: 10.1186/s12902-023-01523-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: 09/03/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023] Open
Abstract
OBJECTIVES Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by chronic hyperglycemia and metabolic stress, involved in the stepwise development of cardiovascular complications. Fibroblast growth factor 21 (FGF21) is a novel hepatokine involved in regulating glucose and lipid metabolism, and has been linked to the prediction, treatment, and improvement of prognosis in multiple cardiovascular diseases (CVDs). The aim of this study is to explore the relationship between FGF21 levels and vascular diseases (VDs) including carotid atherosclerosis (CAS) and hypertension (HP) in patients with T2DM. METHODS Baseline serum FGF21 was determined in a cross-sectional study of 701 patients with T2DM and 258 healthy control. RESULTS The morbidity of CAS was increased in T2DM patients with HP as compared with those without (p < 0.001). The average serum FGF21 level of healthy was [123.9 (67.2-219.3)]. Baseline FGF21 was significantly higher in those who developed CAS or HP than in those who did not [305.9 (177.2-508.4) vs. 197.2 (129.7-308.3) pg/mL, p < 0.001]. In addition, an elevated serum FGF21 was observed in T2DM patients with HP and CAS than that of T2DM patients with CAS or HP [550.5 (312.6-711.3) vs. 305.9 pg/mL, p < 0.001]. Serum FGF21 levels were positively correlated with body mass index and carotid intima media thicknes (p < 0.05), the association remained significant after adjusting for age and T2DM duration. Furthermore, the multinomial logistic regression showed that serum FGF21 was independently associated with CAS and HP in patients with T2DM after adjustment for demographic and traditional VDs risk factors (p < 0.001). CONCLUSIONS Baseline FGF21 is elevated in VDs during diabetes, changes of serum FGF21 levels were appropriately matched to metabolic stress. FGF21can be used as an independent predictor for diagnosing VDs and predicting prognosis.
Collapse
Affiliation(s)
- Zhen Liu
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Yue Peng
- Department of Cardiology, The 1st affiliated Hospital of Wenzhou Medical Unversity, Wenzhou, China
| | - Supeng Li
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Yusheng Lin
- Department of Cardiology, The 1st affiliated Hospital of Wenzhou Medical Unversity, Wenzhou, China
| | - Yunfeng Huang
- Department of Cardiology, The 1st affiliated Hospital of Wenzhou Medical Unversity, Wenzhou, China
| | - Wenting Chen
- Department of Endocrinology, The 3rd affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital, Wenzhou, China
| | - Chunhua Bao
- Department of Endocrinology, The 3rd affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital, Wenzhou, China
| | - Zengxian Zhou
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Zhuofeng Lin
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China.
- Department of Cardiology, The 1st affiliated Hospital of Wenzhou Medical Unversity, Wenzhou, China.
- Laboratory Animal Center of Wenzhou Medical University, Wenzhou, China.
| | - Liangmiao Chen
- Department of Endocrinology, The 3rd affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital, Wenzhou, China.
| |
Collapse
|
29
|
Chen Y, Li Q, Zhao S, Sun L, Yin Z, Wang X, Li X, Iwakiri Y, Han J, Duan Y. Berberine protects mice against type 2 diabetes by promoting PPARγ-FGF21-GLUT2-regulated insulin sensitivity and glucose/lipid homeostasis. Biochem Pharmacol 2023; 218:115928. [PMID: 37979703 DOI: 10.1016/j.bcp.2023.115928] [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: 09/05/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
Type 2 diabetes (T2D) is a chronic, burdensome disease that is characterized by disordered insulin sensitivity and disturbed glucose/lipid homeostasis. Berberine (BBR) has multiple therapeutic actions on T2D, including regulation of glucose and lipid metabolism, improvement of insulin sensitivity and energy expenditure. Recently, the function of BBR on fibroblast growth factor 21 (FGF21) has been identified. However, if BBR ameliorates T2D through FGF21, the underlying mechanisms remain unknown. Herein, we used T2D wild type (WT) and FGF21 global knockout (FKO) mice [mouse T2D model: established by high-fat diet (HFD) feeding plus streptozotocin (STZ) injection], and hepatocyte-specific peroxisome proliferator activated receptor γ (PPARγ) deficient (PPARγHepKO) mice, and cultured human liver carcinoma cells line, HepG2 cells, to characterize the role of BBR in glucose/lipid metabolism and insulin sensitivity. We found that BBR activated FGF21 expression by up-regulating PPARγ expression at the cellular level. Meanwhile, BBR ameliorated glucosamine hydrochloride (Glcn)-induced insulin resistance and increased glucose transporter 2 (GLUT2) expression in a PPARγ/FGF21-dependent manner. In T2D mice, BBR up-regulated the expression of PPARγ, FGF21 and GLUT2 in the liver, and GLUT2 in the pancreas. BBR also reversed T2D-induced insulin resistance, liver lipid accumulation, and damage in liver and pancreas. However, FGF21 deficiency diminished these effects of BBR on diabetic mice. Altogether, our study demonstrates that the therapeutic effects of BBR on T2D were partly accomplished by activating PPARγ-FGF21-GLUT2 signaling pathway. The discovery of this new pathway provides a deeper understanding of the mechanism of BBR for T2D treatment.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cell Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Qi Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Shiwei Zhao
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lei Sun
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Zequn Yin
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaolin Wang
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
30
|
Otani Y, Nozaki Y, Mizunoe Y, Kobayashi M, Higami Y. Effect of mitochondrial quantity and quality controls in white adipose tissue on healthy lifespan: Essential roles of GH/IGF-1-independent pathways in caloric restriction-mediated metabolic remodeling. Pathol Int 2023; 73:479-489. [PMID: 37606202 PMCID: PMC11551837 DOI: 10.1111/pin.13371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023]
Abstract
Long-term caloric restriction is a conventional and reproducible dietary intervention to improve whole body metabolism, suppress age-related pathophysiology, and extend lifespan. The beneficial actions of caloric restriction are widely accepted to be regulated in both growth hormone/insulin-like growth factor 1-dependent and -independent manners. Although growth hormone/insulin-like growth factor 1-dependent regulatory mechanisms are well described, those occurring independent of growth hormone/insulin-like growth factor 1 are poorly understood. In this review, we focus on molecular mechanisms of caloric restriction regulated in a growth hormone/insulin-like growth factor 1-independent manner. Caloric restriction increases mitochondrial quantity and improves mitochondrial quality by activating an axis involving sterol regulatory element binding protein-c/peroxisome proliferator-activated receptor γ coactivator-1α/mitochondrial intermediate peptidase in a growth hormone/insulin-like growth factor 1-independent manner, particularly in white adipose tissue. Fibroblast growth factor 21 is also involved in this axis. Moreover, the axis may be regulated by lower leptin signaling. Thus, caloric restriction appears to induce beneficial actions partially by regulating mitochondrial quantity and quality in white adipose tissue in a growth hormone/insulin-like growth factor 1-independent manner.
Collapse
Grants
- Fostering Joint International Research (B) / 20KK0 Ministry of Education, Culture, Sports, Science and Technology
- Grant-in-Aid for Scientific Research (B) / 17H0217 Ministry of Education, Culture, Sports, Science and Technology
- Grant-in-Aid for Scientific Research (B) / 20H0413 Ministry of Education, Culture, Sports, Science and Technology
- Japan Society for the Promotion of Science Ministry of Education, Culture, Sports, Science and Technology
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Yuina Otani
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical SciencesTokyo University of ScienceChibaJapan
| | - Yuka Nozaki
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical SciencesTokyo University of ScienceChibaJapan
| | - Yuhei Mizunoe
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical SciencesTokyo University of ScienceChibaJapan
| | - Masaki Kobayashi
- Department of Nutrition and Food Science, Graduate School of Humanities and SciencesOchanomizu UniversityTokyoJapan
- Institute for Human Life InnovationOchanomizu UniversityTokyoJapan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical SciencesTokyo University of ScienceChibaJapan
- Research Institute for Biomedical Sciences (RIBS)Tokyo University of ScienceChibaJapan
| |
Collapse
|
31
|
Kobayashi K, Iwasa K, Azuma-Suzuki R, Kawauchi T, Nabeshima YI. Feto-maternal cholesterol transport regulated by β-Klotho-FGF15 axis is essential for fetal growth. Life Sci Alliance 2023; 6:e202301916. [PMID: 37541847 PMCID: PMC10403640 DOI: 10.26508/lsa.202301916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/06/2023] Open
Abstract
β-Klotho (β-KL) is indispensable to regulate lipid, glucose, and energy metabolism in adult animals. β-KL is highly expressed in the yolk sac, but its role in the developmental stages has not been established. We hypothesized that β-KL is required for metabolic regulation in the embryo and aimed to clarify the role of β-KL during development. Here, we show that β-KL regulates feto-maternal cholesterol transport through the yolk sac by mediating FGF 15 signaling, and also that impairment of the β-KL-FGF15 axis causes fetal growth restriction (FGR). Embryos of β- kl knockout (β-kl-/-) mice were morphologically normal but exhibited FGR before placental maturation. The body weight of β-kl-/- mice remained lower after birth. β-KL deletion reduced cholesterol supply from the maternal blood and led to lipid shortage in the embryos. These phenotypes were similar to those of embryos lacking FGF15, indicating that β-KL-FGF15 axis is essential for growth and lipid regulation in the embryonic stages. Our findings suggest that lipid abnormalities in early gestation provoke FGR, leading to reduced body size in later life.
Collapse
Affiliation(s)
- Kanako Kobayashi
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Kazuko Iwasa
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Rika Azuma-Suzuki
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Takeshi Kawauchi
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Adaptive and Maladaptive Responses in Health and Disease, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yo-Ichi Nabeshima
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| |
Collapse
|
32
|
Takebe N, Hasegawa Y, Matsushita Y, Chiba H, Onodera K, Kinno H, Oda T, Nagasawa K, Segawa T, Takahashi Y, Okada K, Ishigaki Y. Association of plasminogen activator inhibitor-1 and fibroblastic growth factor 21 in 3 groups of type 2 diabetes: Without overweight/obesity, free of insulin resistance, and without hepatosteatosis. Medicine (Baltimore) 2023; 102:e34797. [PMID: 37657012 PMCID: PMC10476825 DOI: 10.1097/md.0000000000034797] [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: 05/20/2023] [Accepted: 07/26/2023] [Indexed: 09/03/2023] Open
Abstract
The physiological effects of fibroblast growth factor 21 (FGF21), leading to beneficial metabolic outcomes, have been extensively revealed in recent decades. Significantly elevated serum levels of FGF21 in obesity and type 2 diabetes mellitus (T2DM) are referred to as FGF21 resistance. However, Asian population tend to develop metabolic disorders at a lesser degree of obesity than those of Western. This study aimed to explore factors potentially related to serum FGF21 according to the severity of metabolic disorders in patients with T2DM. This cross-sectional study included 176 T2DM patients. The patients were categorized according to whether they had hepatic steatosis (fatty liver index [FLI] ≥ 60), insulin resistance (homeostasis model assessment of insulin resistance [HOMA-R] ≥ median), and/or overweight/obesity (body mass index [BMI] ≥ 25.0 kg/m2). Independent predictors of serum FGF21 were determined using multiple linear regression analysis in these 3 groups of T2DM patients. Circulating FGF21 levels were correlated positively with BMI, abdominal fat areas, leptin, and plasminogen activator inhibitor-1 (PAI-1). After adjustment for potential confounders, multiple linear regression analysis identified leptin as a factor strongly associated with serum FGF21 levels in all patients. Moreover, PAI-1 was a significant predictor of FGF21 in those with FLI < 60, BMI < 25.0 kg/m2, and HOMA-R < median, while leptin was the only independent factor in each of their counterparts. The factors related to serum FGF21 differ according to the severity of metabolic disorders. FGF21 appears to be independently associated with PAI-1 in T2DM patients: without overweight/obesity, those free of insulin resistance, and those without hepatic steatosis.
Collapse
Affiliation(s)
- Noriko Takebe
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Yutaka Hasegawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Yuriko Matsushita
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Hiraku Chiba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Ken Onodera
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Hirofumi Kinno
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Tomoyasu Oda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Kan Nagasawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Toshie Segawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Yoshihiko Takahashi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Kenta Okada
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Iwate, Japan
| |
Collapse
|
33
|
Kinne AS, Tillman EJ, Abdeen SJ, Johnson DE, Parmer ES, Hurst JP, de Temple B, Rinker S, Rolph TP, Bowsher RR. Noncompetitive immunoassay optimized for pharmacokinetic assessments of biologically active efruxifermin. J Pharm Biomed Anal 2023; 232:115402. [PMID: 37141854 DOI: 10.1016/j.jpba.2023.115402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023]
Abstract
Efruxifermin (EFX) is a homodimeric human IgG1 Fc-FGF21 fusion protein undergoing investigation for treatment of liver fibrosis due to nonalcoholic steatohepatitis (NASH), a prevalent and serious metabolic disease for which there is no approved treatment. Biological activity of FGF21 requires its intact C-terminus, which enables binding to its obligate co-receptor β-Klotho on the surface of target cells. This interaction is a prerequisite for FGF21 signal transduction through its canonical FGF receptors: FGFR1c, 2c, and 3c. Therefore, the C-terminus of each FGF21 polypeptide chain must be intact, with no proteolytic truncation, for EFX to exert its pharmacological activity in patients. A sensitive immunoassay for quantification of biologically active EFX in human serum was therefore needed to support pharmacokinetic assessments in patients with NASH. We present a validated noncompetitive electrochemiluminescent immunoassay (ECLIA) that employs a rat monoclonal antibody for specific capture of EFX via its intact C-terminus. Bound EFX is detected by a SULFO-TAG™-conjugated, affinity purified chicken anti-EFX antiserum. The ECLIA reported herein for quantification of EFX demonstrated suitable analytical performance, with a sensitivity (LLOQ) of 20.0 ng/mL, to support reliable pharmacokinetic assessments of EFX. The validated assay was used to quantify serum EFX concentrations in a phase 2a study of NASH patients (BALANCED) with either moderate-to-advanced fibrosis or compensated cirrhosis. The pharmacokinetic profile of EFX was dose-proportional and did not differ between patients with moderate-to-advanced fibrosis and those with compensated cirrhosis. This report presents the first example of a validated pharmacokinetic assay specific for a biologically active Fc-FGF21 fusion protein, as well as the first demonstration of use of a chicken antibody conjugate as a detection reagent specific for an FGF21 analog.
Collapse
Affiliation(s)
- Adam S Kinne
- B2S Life Sciences LLC, 97 East Monroe Street, Franklin, IN 46131, USA
| | - Erik J Tillman
- Akero Therapeutics, 601 Gateway Blvd. #350, South San Francisco, CA 94080, USA
| | - Sanofar J Abdeen
- B2S Life Sciences LLC, 97 East Monroe Street, Franklin, IN 46131, USA
| | - Derrick E Johnson
- B2S Life Sciences LLC, 97 East Monroe Street, Franklin, IN 46131, USA
| | - Elijah S Parmer
- B2S Life Sciences LLC, 97 East Monroe Street, Franklin, IN 46131, USA
| | - Jacob P Hurst
- B2S Life Sciences LLC, 97 East Monroe Street, Franklin, IN 46131, USA
| | - Brittany de Temple
- Akero Therapeutics, 601 Gateway Blvd. #350, South San Francisco, CA 94080, USA
| | - Sherri Rinker
- B2S Life Sciences LLC, 97 East Monroe Street, Franklin, IN 46131, USA
| | - Timothy P Rolph
- Akero Therapeutics, 601 Gateway Blvd. #350, South San Francisco, CA 94080, USA
| | - Ronald R Bowsher
- B2S Life Sciences LLC, 97 East Monroe Street, Franklin, IN 46131, USA.
| |
Collapse
|
34
|
Le TDV, Fathi P, Watters AB, Ellis BJ, Besing GLK, Bozadjieva-Kramer N, Perez MB, Sullivan AI, Rose JP, Baggio LL, Koehler J, Brown JL, Bales MB, Nwaba KG, Campbell JE, Drucker DJ, Potthoff MJ, Seeley RJ, Ayala JE. Fibroblast growth factor-21 is required for weight loss induced by the glucagon-like peptide-1 receptor agonist liraglutide in male mice fed high carbohydrate diets. Mol Metab 2023; 72:101718. [PMID: 37030441 PMCID: PMC10131131 DOI: 10.1016/j.molmet.2023.101718] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
OBJECTIVE Glucagon-like peptide-1 receptor (GLP-1R) agonists (GLP-1RA) and fibroblast growth factor-21 (FGF21) confer similar metabolic benefits. GLP-1RA induce FGF21, leading us to investigate mechanisms engaged by the GLP-1RA liraglutide to increase FGF21 levels and the metabolic relevance of liraglutide-induced FGF21. METHODS Circulating FGF21 levels were measured in fasted male C57BL/6J, neuronal GLP-1R knockout, β-cell GLP-1R knockout, and liver peroxisome proliferator-activated receptor alpha knockout mice treated acutely with liraglutide. To test the metabolic relevance of liver FGF21 in response to liraglutide, chow-fed control and liver Fgf21 knockout (LivFgf21-/-) mice were treated with vehicle or liraglutide in metabolic chambers. Body weight and composition, food intake, and energy expenditure were measured. Since FGF21 reduces carbohydrate intake, we measured body weight in mice fed matched diets with low- (LC) or high-carbohydrate (HC) content and in mice fed a high-fat, high-sugar (HFHS) diet. This was done in control and LivFgf21-/- mice and in mice lacking neuronal β-klotho (Klb) expression to disrupt brain FGF21 signaling. RESULTS Liraglutide increases FGF21 levels independently of decreased food intake via neuronal GLP-1R activation. Lack of liver Fgf21 expression confers resistance to liraglutide-induced weight loss due to attenuated reduction of food intake in chow-fed mice. Liraglutide-induced weight loss was impaired in LivFgf21-/- mice when fed HC and HFHS diets but not when fed a LC diet. Loss of neuronal Klb also attenuated liraglutide-induced weight loss in mice fed HC or HFHS diets. CONCLUSIONS Our findings support a novel role for a GLP-1R-FGF21 axis in regulating body weight in a dietary carbohydrate-dependent manner.
Collapse
Affiliation(s)
- Thao D V Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Payam Fathi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Amanda B Watters
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Blair J Ellis
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Gai-Linn K Besing
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Veterans Affairs Ann Arbor Healthcare System, Research Service, 2215 Fuller Road, Ann Arbor, MI 48105, USA.
| | - Misty B Perez
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Andrew I Sullivan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Jesse P Rose
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Laurie L Baggio
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| | - Jacqueline Koehler
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | - Jennifer L Brown
- Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, Durham, NC 27701, USA
| | - Michelle B Bales
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Kaitlyn G Nwaba
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, Durham, NC 27701, USA.
| | - Daniel J Drucker
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Randy J Seeley
- Department of Surgery, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA.
| | - Julio E Ayala
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
35
|
Aaldijk AS, Verzijl CRC, Jonker JW, Struik D. Biological and pharmacological functions of the FGF19- and FGF21-coreceptor beta klotho. Front Endocrinol (Lausanne) 2023; 14:1150222. [PMID: 37260446 PMCID: PMC10229096 DOI: 10.3389/fendo.2023.1150222] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/13/2023] [Indexed: 06/02/2023] Open
Abstract
Beta klotho (KLB) is a fundamental component in fibroblast growth factor receptor (FGFR) signaling as it serves as an obligatory coreceptor for the endocrine hormones fibroblast growth factor 19 (FGF19) and fibroblast growth factor 21 (FGF21). Through the development of FGF19- and FGF21 mimetics, KLB has emerged as a promising drug target for treating various metabolic diseases, such as type 2 diabetes (T2D), non-alcoholic fatty liver disease (NAFLD), and cardiovascular disease. While rodent studies have significantly increased our understanding of KLB function, current clinical trials that test the safety and efficacy of KLB-targeting drugs raise many new scientific questions about human KLB biology. Although most KLB-targeting drugs can modulate disease activity in humans, individual patient responses differ substantially. In addition, species-specific differences in KLB tissue distribution may explain why the glucose-lowering effects that were observed in preclinical studies are not fully replicated in clinical trials. Besides, the long-term efficacy of KLB-targeting drugs might be limited by various pathophysiological conditions known to reduce the expression of KLB. Moreover, FGF19/FGF21 administration in humans is also associated with gastrointestinal side effects, which are currently unexplained. A better understanding of human KLB biology could help to improve the efficacy and safety of existing or novel KLB/FGFR-targeting drugs. In this review, we provide a comprehensive overview of the current understanding of KLB biology, including genetic variants and their phenotypic associations, transcriptional regulation, protein structure, tissue distribution, subcellular localization, and function. In addition, we will highlight recent developments regarding the safety and efficacy of KLB-targeting drugs in clinical trials. These insights may direct the development and testing of existing and future KLB-targeting drugs.
Collapse
|
36
|
Goutam RS, Kumar V, Lee U, Kim J. Exploring the Structural and Functional Diversity among FGF Signals: A Comparative Study of Human, Mouse, and Xenopus FGF Ligands in Embryonic Development and Cancer Pathogenesis. Int J Mol Sci 2023; 24:ijms24087556. [PMID: 37108717 PMCID: PMC10146080 DOI: 10.3390/ijms24087556] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Fibroblast growth factors (FGFs) encode a large family of growth factor proteins that activate several intracellular signaling pathways to control diverse physiological functions. The human genome encodes 22 FGFs that share a high sequence and structural homology with those of other vertebrates. FGFs orchestrate diverse biological functions by regulating cellular differentiation, proliferation, and migration. Dysregulated FGF signaling may contribute to several pathological conditions, including cancer. Notably, FGFs exhibit wide functional diversity among different vertebrates spatiotemporally. A comparative study of FGF receptor ligands and their diverse roles in vertebrates ranging from embryonic development to pathological conditions may expand our understanding of FGF. Moreover, targeting diverse FGF signals requires knowledge regarding their structural and functional heterogeneity among vertebrates. This study summarizes the current understanding of human FGF signals and correlates them with those in mouse and Xenopus models, thereby facilitating the identification of therapeutic targets for various human disorders.
Collapse
Affiliation(s)
- Ravi Shankar Goutam
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- iPS Bio, Inc., 3F, 16 Daewangpangyo-ro 712 Beon-gil, Bundang-gu, Seongnam-si 13522, Republic of Korea
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
37
|
Wang H, Xiong Q, He G, Tang J, Sun L, Cheng S, Ke M, Chen S, Hu Y, Feng J, Song L, Tong B, Zhang Z, Dai Z, Xu Y. Hepatic IDH2 regulates glycolysis and gluconeogenesis. Metabolism 2023; 143:155559. [PMID: 37044373 DOI: 10.1016/j.metabol.2023.155559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND AND AIMS The liver plays a central role in controlling glucose and lipid metabolism. IDH2, a mitochondrial protein, controls TCA cycle flux. However, its role in regulating metabolism in obesity is still unclear. This study intends to investigate the impact of hepatic IDH2 expression on overnutrition-regulated glucose and lipid metabolism. METHODS Hepatic IDH2 was knocked-out in mice by the approach of CRISPR-Cas9. Mice were subjected to starvation and refeeding for hepatic glucose and lipid studies in vivo. Primary hepatocytes and mouse normal liver cell line, AML12 cells were used for experiments in vitro. RESULTS This study found that IDH2 protein levels were elevated in the livers of obese people and mice with high-fat diet consumption or hepatic steatosis. Liver IDH2-deletion mice (IDH2LKO) were resistant to high-fat diet-induced body weight gain, with lower serum glucose and TG levels, increased insulin sensitivity, and higher FGF21 secretion, despite the higher TG content in the liver. Consistently, overexpression of IDH2 in hepatocytes promoted gluconeogenesis and enhanced glycogenesis. By performing mass spectrometry and proteomics analyses, we further demonstrated that IDH2-deficiency in hepatocytes accelerated ATP production by increasing forward TCA cycle flux, thus promoting glycolysis pathway and decreasing glycogen synthesis at refeeding state, and inhibiting hepatic gluconeogenesis, increasing β-oxidation during starvation. Moreover, experiments in vivo demonstrated that IDH2-knockout might not exacerbate hepatic inflammatory responses in the NASH model. CONCLUSIONS Elevated hepatic IDH2 under over-nutrition state contributes to elevated gluconeogenesis and glycogen synthesis. Inhibition of IDH2 in the liver could be a potential therapeutic target for obesity and diabetes.
Collapse
Affiliation(s)
- Huawei Wang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qing Xiong
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Endocrinology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, China
| | - Guangzhen He
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan 442000, China
| | - Jun Tang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Li Sun
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Siyuan Cheng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Nuclear Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou 510000, China
| | - Mengting Ke
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Biochemistry, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Shangyu Chen
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yong Hu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jieyuan Feng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Linyang Song
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Beier Tong
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhengwei Zhang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhe Dai
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
38
|
Tian H, Zhang S, Liu Y, Wu Y, Zhang D. Fibroblast Growth Factors for Nonalcoholic Fatty Liver Disease: Opportunities and Challenges. Int J Mol Sci 2023; 24:ijms24054583. [PMID: 36902015 PMCID: PMC10003526 DOI: 10.3390/ijms24054583] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), a chronic condition associated with metabolic dysfunction and obesity, has reached epidemic proportions worldwide. Although early NAFLD can be treated with lifestyle changes, the treatment of advanced liver pathology, such as nonalcoholic steatohepatitis (NASH), remains a challenge. There are currently no FDA-approved drugs for NAFLD. Fibroblast growth factors (FGFs) play essential roles in lipid and carbohydrate metabolism and have recently emerged as promising therapeutic agents for metabolic diseases. Among them, endocrine members (FGF19 and FGF21) and classical members (FGF1 and FGF4) are key regulators of energy metabolism. FGF-based therapies have shown therapeutic benefits in patients with NAFLD, and substantial progress has recently been made in clinical trials. These FGF analogs are effective in alleviating steatosis, liver inflammation, and fibrosis. In this review, we describe the biology of four metabolism-related FGFs (FGF19, FGF21, FGF1, and FGF4) and their basic action mechanisms, and then summarize recent advances in the biopharmaceutical development of FGF-based therapies for patients with NAFLD.
Collapse
Affiliation(s)
- Haoyu Tian
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Shuairan Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ying Liu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Yifan Wu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Dianbao Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
- Correspondence: or
| |
Collapse
|
39
|
Sarkar A, Jin Y, DeFelice BC, Logan CY, Yang Y, Anbarchian T, Wu P, Morri M, Neff NF, Nguyen H, Rulifson E, Fish M, Kaye AG, Martínez Jaimes AM, Nusse R. Intermittent fasting induces rapid hepatocyte proliferation to restore the hepatostat in the mouse liver. eLife 2023; 12:e82311. [PMID: 36719070 PMCID: PMC9889086 DOI: 10.7554/elife.82311] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/09/2022] [Indexed: 02/01/2023] Open
Abstract
Nutrient availability fluctuates in most natural populations, forcing organisms to undergo periods of fasting and re-feeding. It is unknown how dietary changes influence liver homeostasis. Here, we show that a switch from ad libitum feeding to intermittent fasting (IF) promotes rapid hepatocyte proliferation. Mechanistically, IF-induced hepatocyte proliferation is driven by the combined action of systemic FGF15 and localized WNT signaling. Hepatocyte proliferation during periods of fasting and re-feeding re-establishes a constant liver-to-body mass ratio, thus maintaining the hepatostat. This study provides the first example of dietary influence on adult hepatocyte proliferation and challenges the widely held view that liver tissue is mostly quiescent unless chemically or mechanically injured.
Collapse
Affiliation(s)
- Abby Sarkar
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Yinhua Jin
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | | | - Catriona Y Logan
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Yan Yang
- Stanford Center for Genomics & Personalized Medicine, Stanford University School of MedicineStanfordUnited States
| | - Teni Anbarchian
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Peng Wu
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | | | - Norma F Neff
- Chan-Zuckerberg BiohubSan FranciscoUnited States
| | - Huy Nguyen
- Department of Neurology and Neurological Sciences, Stanford University School of MedicineStanfordUnited States
| | - Eric Rulifson
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Matthew Fish
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Avi Gurion Kaye
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Azalia M Martínez Jaimes
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Roel Nusse
- Howard Hughes Medical Institute, Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
40
|
Jin L, Yang R, Geng L, Xu A. Fibroblast Growth Factor-Based Pharmacotherapies for the Treatment of Obesity-Related Metabolic Complications. Annu Rev Pharmacol Toxicol 2023; 63:359-382. [PMID: 36100222 DOI: 10.1146/annurev-pharmtox-032322-093904] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The fibroblast growth factor (FGF) family, which comprises 22 structurally related proteins, plays diverse roles in cell proliferation, differentiation, development, and metabolism. Among them, two classical members (FGF1 and FGF4) and two endocrine members (FGF19 and FGF21) are important regulators of whole-body energy homeostasis, glucose/lipid metabolism, and insulin sensitivity. Preclinical studies have consistently demonstrated the therapeutic benefits of these FGFs for the treatment of obesity, diabetes, dyslipidemia, and nonalcoholic steatohepatitis (NASH). Several genetically engineered FGF19 and FGF21 analogs with improved pharmacodynamic and pharmacokinetic properties have been developed and progressed into various stages of clinical trials. These FGF analogs are effective in alleviating hepatic steatosis, steatohepatitis, and liver fibrosis in biopsy-confirmed NASH patients, whereas their antidiabetic and antiobesity effects are mildand vary greatly in different clinical trials. This review summarizes recent advances in biopharmaceutical development of FGF-based therapies against obesity-related metabolic complications, highlights major challenges in clinical implementation, and discusses possible strategies to overcome these hurdles.
Collapse
Affiliation(s)
- Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ranyao Yang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Leiluo Geng
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
41
|
Huang W, Zhang J, Zou J, Wang X, Xu H, Guan J, Yi H, Liu S, Yin S. Fibroblast growth factor 21 is an independent predictor of prevalent and incident obstructive sleep apnea. iScience 2023; 26:105985. [PMID: 36798439 PMCID: PMC9926096 DOI: 10.1016/j.isci.2023.105985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/29/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a metabolic regulator and a potential biomarker of metabolic diseases. Limited data are available on the association between FGF21 and obstructive sleep apnea (OSA), which is considered as a manifestation of metabolic syndrome. In the present cross-sectional and longitudinal analyses, the FGF21 level was associated with OSA. This analysis of two clinical cohorts is the first to show that the FGF21 level increased significantly with OSA severity and was an independent predictor of incident OSA in Chinese adults. The circulating FGF21 level could serve as a potential serum biomarker of OSA and its comorbidities and thus aid risk evaluation and early intervention.
Collapse
Affiliation(s)
- Weijun Huang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China
| | - Jingyu Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China
| | - Jianyin Zou
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China
| | - Xiaoting Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China
| | - Huajun Xu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China
| | - Jian Guan
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China
| | - Hongliang Yi
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China,Corresponding author
| | - Suru Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China,Corresponding author
| | - Shankai Yin
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, China,Shanghai Key Laboratory of Sleep Disordered Breathing, 600 Yishan Road, Shanghai, China,Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, China
| |
Collapse
|
42
|
Le TDV, Fathi P, Watters AB, Ellis BJ, Bozadjieva-Kramer N, Perez MB, Sullivan AI, Rose JP, Baggio LL, Koehler J, Brown JL, Bales MB, Nwaba KG, Campbell JE, Drucker DJ, Potthoff MJ, Seeley RJ, Ayala JE. Liver Fibroblast Growth Factor 21 (FGF21) is Required for the Full Anorectic Effect of the Glucagon-Like Peptide-1 Receptor Agonist Liraglutide in Male Mice fed High Carbohydrate Diets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522509. [PMID: 36711605 PMCID: PMC9881863 DOI: 10.1101/2023.01.03.522509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists and fibroblast growth factor 21 (FGF21) confer similar metabolic benefits. Studies report that GLP-1RA induce FGF21. Here, we investigated the mechanisms engaged by the GLP-1R agonist liraglutide to increase FGF21 levels and the metabolic relevance of liraglutide-induced FGF21. We show that liraglutide increases FGF21 levels via neuronal GLP-1R activation. We also demonstrate that lack of liver Fgf21 expression confers partial resistance to liraglutide-induced weight loss. Since FGF21 reduces carbohydrate intake, we tested whether the contribution of FGF21 to liraglutide-induced weight loss is dependent on dietary carbohydrate content. In control and liver Fgf21 knockout (Liv Fgf21 -/- ) mice fed calorically matched diets with low- (LC) or high-carbohydrate (HC) content, we found that only HC-fed Liv Fgf21 -/- mice were resistant to liraglutide-induced weight loss. Similarly, liraglutide-induced weight loss was partially impaired in Liv Fgf21 -/- mice fed a high-fat, high-sugar (HFHS) diet. Lastly, we show that loss of neuronal β-klotho expression also diminishes liraglutide-induced weight loss in mice fed a HC or HFHS diet, indicating that FGF21 mediates liraglutide-induced weight loss via neuronal FGF21 action. Our findings support a novel role for a GLP-1R-FGF21 axis in regulating body weight in the presence of high dietary carbohydrate content.
Collapse
|
43
|
Akduman F, Şıklar Z, Özsu E, Doğan Ö, Kır MK, Berberoğlu M. Fibroblast Growth Factor 21 Levels and Bone Mineral Density in Metabolically Healthy and Metabolically Unhealthy Obese Children. J Clin Res Pediatr Endocrinol 2022; 14:433-443. [PMID: 35859690 PMCID: PMC9724058 DOI: 10.4274/jcrpe.galenos.2022.2022-1-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The harmful or beneficial effect of obesity on bone mineral density (BMD) remains controversial in children and adolescents. Fibroblast growth factor 21 (FGF21) is a metabolic factor that plays a specific role in the regulation of carbohydrate and lipid metabolism. However, the role of FGF21 in bone metabolism appears paradoxical and is complex. To determine whether serum FGF21 level was associated with BMD in obese children and adolescents. METHODS The study was conducted with the participation of children and adolescents aged 8-18 years. Ninety-eight obese children were included in the study group and 44 children were included in the control group. BMD, in addition to the routine obesity workup, which includes fasting blood glucose, fasting insulin levels, lipid profile, and liver enzymes; serum FGF21 levels have been analyzed. RESULTS The mean age of the obese group (n=98) was 13.34±2.24 years and the mean age of controls (n=44) was 13.48±2.87 years. Based on International Diabetes Federation criteria, 15 of 98 (15.3%) patients were metabolically unhealthy. FGF21 levels were 193.54±139.62 mg/dL in the obese group and 158.69±151.81 mg/dL in the control group (p=0.06). There was no difference between the FGF21 and BMD z-score values of girls and boys in the obese and control groups (p>0.05). CONCLUSION BMD-z-score was increased in obese children compared to healthy control. Moreover, BMD-z-score tended to be higher when more metabolic risk factors were present. However, there was no significant relationship between FGF21 levels and BMD z-score values in obese children.
Collapse
Affiliation(s)
- Filiz Akduman
- Ankara University Faculty of Medicine, Department of Pediatrics, Ankara, Turkey,* Address for Correspondence: Ankara University Faculty of Medicine, Department of Pediatrics, Ankara, Turkey Phone: +90 507 443 24 42 E-mail:
| | - Zeynep Şıklar
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Elif Özsu
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Özlem Doğan
- Ankara University Faculty of Medicine, Department of Clinical Biochemistry, Ankara, Turkey
| | - Metin Kemal Kır
- Ankara University School of Medicine, Department of Nuclear Medicine, Ankara, Turkey
| | - Merih Berberoğlu
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey
| |
Collapse
|
44
|
Prida E, Álvarez-Delgado S, Pérez-Lois R, Soto-Tielas M, Estany-Gestal A, Fernø J, Seoane LM, Quiñones M, Al-Massadi O. Liver Brain Interactions: Focus on FGF21 a Systematic Review. Int J Mol Sci 2022; 23:ijms232113318. [PMID: 36362103 PMCID: PMC9658462 DOI: 10.3390/ijms232113318] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/21/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor 21 is a pleiotropic hormone secreted mainly by the liver in response to metabolic and nutritional challenges. Physiologically, fibroblast growth factor 21 plays a key role in mediating the metabolic responses to fasting or starvation and acts as an important regulator of energy homeostasis, glucose and lipid metabolism, and insulin sensitivity, in part by its direct action on the central nervous system. Accordingly, pharmacological recombinant fibroblast growth factor 21 therapies have been shown to counteract obesity and its related metabolic disorders in both rodents and nonhuman primates. In this systematic review, we discuss how fibroblast growth factor 21 regulates metabolism and its interactions with the central nervous system. In addition, we also state our vision for possible therapeutic uses of this hepatic-brain axis.
Collapse
Affiliation(s)
- Eva Prida
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Sara Álvarez-Delgado
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Raquel Pérez-Lois
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
| | - Mateo Soto-Tielas
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Unidad de Metodología de la Investigación, Fundación Instituto de Investigación de Santiago (FIDIS), 15706 Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, 5201 Bergen, Norway
| | - Luisa María Seoane
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
| | - Mar Quiñones
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
- Correspondence: (M.Q.); (O.A.-M.); Tel.: +34-981955708 (M.Q.); +34-981955522 (O.A.-M.)
| | - Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
- Correspondence: (M.Q.); (O.A.-M.); Tel.: +34-981955708 (M.Q.); +34-981955522 (O.A.-M.)
| |
Collapse
|
45
|
Patel S, Haider A, Alvarez-Guaita A, Bidault G, El-Sayed Moustafa JS, Guiu-Jurado E, Tadross JA, Warner J, Harrison J, Virtue S, Scurria F, Zvetkova I, Blüher M, Small KS, O'Rahilly S, Savage DB. Combined genetic deletion of GDF15 and FGF21 has modest effects on body weight, hepatic steatosis and insulin resistance in high fat fed mice. Mol Metab 2022; 65:101589. [PMID: 36064109 PMCID: PMC9486046 DOI: 10.1016/j.molmet.2022.101589] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES Obesity in humans and mice is associated with elevated levels of two hormones responsive to cellular stress, namely GDF15 and FGF21. Over-expression of each of these is associated with weight loss and beneficial metabolic changes but where they are secreted from and what they are required for physiologically in the context of overfeeding remains unclear. METHODS Here we used tissue selective knockout mouse models and human transcriptomics to determine the source of circulating GDF15 in obesity. We then generated and characterized the metabolic phenotypes of GDF15/FGF21 double knockout mice. RESULTS Circulating GDF15 and FGF21 are both largely derived from the liver, rather than adipose tissue or skeletal muscle, in obese states. Combined whole body deletion of FGF21 and GDF15 does not result in any additional weight gain in response to high fat feeding but it does result in significantly greater hepatic steatosis and insulin resistance than that seen in GDF15 single knockout mice. CONCLUSIONS Collectively the data suggest that overfeeding activates a stress response in the liver which is the major source of systemic rises in GDF15 and FGF21. These hormones then activate pathways which reduce this metabolic stress.
Collapse
Affiliation(s)
- Satish Patel
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK; MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | - Afreen Haider
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK; MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | - Anna Alvarez-Guaita
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | | | - Esther Guiu-Jurado
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - John A Tadross
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK; East Midlands and East of England Genomic Laboratory Hub & Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James Warner
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - James Harrison
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Samuel Virtue
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - Fabio Scurria
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - Ilona Zvetkova
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, SE1 7EH, UK
| | - Stephen O'Rahilly
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK; MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - David B Savage
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK; MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
46
|
Nies VJM, Struik D, Liu S, Liu W, Kruit JK, Downes M, van Zutphen T, Verkade HJ, Evans RM, Jonker JW. Autocrine FGF1 signaling promotes glucose uptake in adipocytes. Proc Natl Acad Sci U S A 2022; 119:e2122382119. [PMID: 36161959 PMCID: PMC9546606 DOI: 10.1073/pnas.2122382119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor 1 (FGF1) is an autocrine growth factor released from adipose tissue during over-nutrition or fasting to feeding transition. While local actions underlie the majority of FGF1's anti-diabetic functions, the molecular mechanisms downstream of adipose FGF receptor signaling are unclear. We investigated the effects of FGF1 on glucose uptake and its underlying mechanism in murine 3T3-L1 adipocytes and in ex vivo adipose explants from mice. FGF1 increased glucose uptake in 3T3-L1 adipocytes and epididymal WAT (eWAT) and inguinal WAT (iWAT). Conversely, glucose uptake was reduced in eWAT and iWAT of FGF1 knockout mice. We show that FGF1 acutely increased adipocyte glucose uptake via activation of the insulin-sensitive glucose transporter GLUT4, involving dynamic crosstalk between the MEK1/2 and Akt signaling proteins. Prolonged exposure to FGF1 stimulated adipocyte glucose uptake by MEK1/2-dependent transcription of the basal glucose transporter GLUT1. We have thus identified an alternative pathway to stimulate glucose uptake in adipocytes, independent from insulin, which could open new avenues for treating patients with type 2 diabetes.
Collapse
Affiliation(s)
- Vera J. M. Nies
- Laboratory of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Dicky Struik
- Laboratory of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Sihao Liu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Weilin Liu
- Laboratory of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Janine K. Kruit
- Laboratory of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Tim van Zutphen
- Laboratory of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Henkjan J. Verkade
- Laboratory of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Johan W. Jonker
- Laboratory of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
47
|
SUI Y, CHEN J. Hepatic FGF21: Its Emerging Role in Inter-Organ Crosstalk and Cancers. Int J Biol Sci 2022; 18:5928-5942. [PMID: 36263162 PMCID: PMC9576513 DOI: 10.7150/ijbs.76924] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/18/2022] [Indexed: 02/07/2023] Open
Abstract
Fibroblast growth factor (FGF) 21 is one of the FGF members with special endocrine properties. In the last twenty years, it has attracted intense research and development for its physiological functions that respond to dietary manipulation, pharmacological benefits of improving the macronutrient metabolism, and clinical values as a biomarker of various human diseases. Generally, FGF21 can be produced by major metabolic organs, but only the subgroup from the liver shows canonical endocrine properties, which emphasizes the special value of delineating the unique secretory and functional characteristics of hepatic FGF21. There has been a growth in literature to address the extra-hepatic activities of FGF21, and many striking findings have therefore been published. Yet, they are fragmented and scattered, and controversies are raised from divergent findings. For this reason, there is a need for a systematic and critical evaluation of current research in this aspect. In this review, we focus on the current knowledge about the molecular biology of endocrine FGF21, especially present details on the regulation of circulating levels of FGF21. We also emphasize its emerging roles in inter-organ crosstalk and cancer development.
Collapse
Affiliation(s)
- Yue SUI
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jianping CHEN
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
48
|
Claflin KE, Sullivan AI, Naber MC, Flippo KH, Morgan DA, Neff TJ, Jensen-Cody SO, Zhu Z, Zingman LV, Rahmouni K, Potthoff MJ. Pharmacological FGF21 signals to glutamatergic neurons to enhance leptin action and lower body weight during obesity. Mol Metab 2022; 64:101564. [PMID: 35944896 PMCID: PMC9403559 DOI: 10.1016/j.molmet.2022.101564] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Fibroblast growth factor 21 (FGF21) is a peripherally-derived endocrine hormone that acts on the central nervous system (CNS) to regulate whole body energy homeostasis. Pharmacological administration of FGF21 promotes weight loss in obese animal models and human subjects with obesity. However, the central targets mediating these effects are incompletely defined. METHODS To explore the mechanism for FGF21's effects to lower body weight, we pharmacologically administer FGF21 to genetic animal models lacking the obligate FGF21 co-receptor, β-klotho (KLB), in either glutamatergic (Vglut2-Cre) or GABAergic (Vgat-Cre) neurons. In addition, we abolish FGF21 signaling to leptin receptor (LepR-Cre) positive cells. Finally, we examine the synergistic effects of FGF21 and leptin to lower body weight and explore the importance of physiological leptin levels in FGF21-mediated regulation of body weight. RESULTS Here we show that FGF21 signaling to glutamatergic neurons is required for FGF21 to modulate energy expenditure and promote weight loss. In addition, we demonstrate that FGF21 signals to leptin receptor-expressing cells to regulate body weight, and that central leptin signaling is required for FGF21 to fully stimulate body weight loss during obesity. Interestingly, co-administration of FGF21 and leptin synergistically leads to robust weight loss. CONCLUSIONS These data reveal an important endocrine crosstalk between liver- and adipose-derived signals which integrate in the CNS to modulate energy homeostasis and body weight regulation.
Collapse
Affiliation(s)
- Kristin E Claflin
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andrew I Sullivan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Meghan C Naber
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kyle H Flippo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Tate J Neff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Sharon O Jensen-Cody
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zhiyong Zhu
- Department of Internal Medicine, Iowa City, IA 52242, USA
| | | | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Veterans Affairs Health Care System, Iowa City, IA 52242, USA; Department of Internal Medicine, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Veterans Affairs Health Care System, Iowa City, IA 52242, USA.
| |
Collapse
|
49
|
She QY, Li LJ, Liu MH, Tan RY, Zhong YW, Bao JF, Xie JD. Bibliometric analysis of fibroblast growth factor 21 research over the period 2000 to 2021. Front Pharmacol 2022; 13:1011008. [PMID: 36238554 PMCID: PMC9551462 DOI: 10.3389/fphar.2022.1011008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 01/10/2023] Open
Abstract
Background: Fibroblast growth factor 21 (FGF-21) is an evolutionarily conserved protein that plays multiple roles in metabolic regulation. Over the past two decades, numerous studies have deepened our understanding of its various functions and its pharmacological value. Nevertheless, most clinical trials have not achieved the desired results, which raises issues regarding its clinical value. In this bibliometric analysis, we evaluated the state of FGF-21 research over the last 20 years and identified important topics, achievements, and potential future directions. Methods: Publications related to FGF-21 were collected from the Web of Science Core Collection-Science Citation Index Expanded. HistCite, VOSviewer, and CiteSpace were used for bibliometric analysis and visualization, including the analysis of annual publications, leading countries, active institutions and authors, core journals, co-cited references, and keywords. Results: Altogether, 2,490 publications related to FGF-21 were obtained. A total of 12,872 authors from 2,628 institutions in 77 countries or regions reported studies on FGF-21. The United States of America was the most influential country in FGF-21 research. Alexei Kharitonenkov, Steven A. Kliewer, and David J. Mangelsdorf were the most influential scholars, and endocrinology journals had a core status in the field. The physiological roles, clinical translation, and FGF-21-based drug development were the main topics of research, and future studies may concentrate on the central effects of FGF-21, FGF-21-based drug development, and the effects of FGF-21 on non-metabolic diseases. Conclusion: The peripheral metabolic effects of FGF-21, FGF-21-based drug development, and translational research on metabolic diseases are the three major topics in FGF-21 research, whereas the central metabolic effects of FGF-21 and the effects of FGF-21 on metabolic diseases are the emerging trends and may become the following hot topics in FGF-21 research.
Collapse
Affiliation(s)
- Qin-Ying She
- Department of Nephrology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Li-Juan Li
- Department of Nephrology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Ming-Hong Liu
- Department of Nephrology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Ru-Yu Tan
- Department of Nephrology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yi-Wen Zhong
- Department of Nephrology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Fu Bao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jie-Dong Xie
- Department of Nephrology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Jie-Dong Xie,
| |
Collapse
|
50
|
Ayana G, Ryu J, Choe SW. Ultrasound-Responsive Nanocarriers for Breast Cancer Chemotherapy. MICROMACHINES 2022; 13:1508. [PMID: 36144131 PMCID: PMC9503784 DOI: 10.3390/mi13091508] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 05/13/2023]
Abstract
Breast cancer is the most common type of cancer and it is treated with surgical intervention, radiotherapy, chemotherapy, or a combination of these regimens. Despite chemotherapy's ample use, it has limitations such as bioavailability, adverse side effects, high-dose requirements, low therapeutic indices, multiple drug resistance development, and non-specific targeting. Drug delivery vehicles or carriers, of which nanocarriers are prominent, have been introduced to overcome chemotherapy limitations. Nanocarriers have been preferentially used in breast cancer chemotherapy because of their role in protecting therapeutic agents from degradation, enabling efficient drug concentration in target cells or tissues, overcoming drug resistance, and their relatively small size. However, nanocarriers are affected by physiological barriers, bioavailability of transported drugs, and other factors. To resolve these issues, the use of external stimuli has been introduced, such as ultrasound, infrared light, thermal stimulation, microwaves, and X-rays. Recently, ultrasound-responsive nanocarriers have become popular because they are cost-effective, non-invasive, specific, tissue-penetrating, and deliver high drug concentrations to their target. In this paper, we review recent developments in ultrasound-guided nanocarriers for breast cancer chemotherapy, discuss the relevant challenges, and provide insights into future directions.
Collapse
Affiliation(s)
- Gelan Ayana
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| | - Jaemyung Ryu
- Department of Optical Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| | - Se-woon Choe
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| |
Collapse
|