1
|
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
|
2
|
Lee B, Jo D, Park J, Kim OY, Song J. Gut microbiota and their relationship with circulating adipokines in an acute hepatic encephalopathy mouse model induced by surgical bile duct ligation. Heliyon 2024; 10:e38534. [PMID: 39391493 PMCID: PMC11466606 DOI: 10.1016/j.heliyon.2024.e38534] [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: 06/18/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Background and aims Various studies have shown the importance of the gut microbiota in human health. However, little is known about gut microbiome patterns and their effect on circulating adipo-myokine levels in hepatic encephalopathy (HE). We investigated the relationship between the gut microbiota and adipo-myokine levels using a mouse model of HE induced by surgical bile duct ligation (BDL). Methods and results Wild-type C57BL/6J mice were subjected to sham surgery or BDL. Severe body weight loss, suppressed feed intake, and liver failure were observed in BDL mice compared with sham control mice. Additionally, changes in gut microbial communities and serum adipo-myokine levels were noted in BDL mice. In the BDL mouse gut, we identified 15 differentially abundant taxa including the phylum Verrucomicrobiota, the classes Actinomycetes and Verrucomicrobiae, the order Verrucomicrobiales, the families Akkermansiaceae, Bacteroidaceae, Rikenellaceae, and Oscillospiraceae, the genera Alistipes, Akkermansia, Muribaculum, and Phocaeicola, and the species Akkermansia muciniphila, Alistipes okayasuensis, and Muribaculum gordoncarteri by LEfSe analysis (LDA score≥4.0). Higher levels of certain adipo-myokines such as BDNF were detected in the serum of BDL mice. Spearman correlation analysis revealed that certain adipo-myokines (e.g., FSTL1) were positively correlated with the class Actinomycetes, the family Rikenellaceae, the genus Alistipes, and the species Alistipes okayasuensis. Interestingly, A. okayasuensis and M. gordoncarteri, recently isolated microbes, showed richness in the gut of BDL mice and demonstrated positive correlations with adipo-myokines such as FGF21. Conclusions Overall, our results suggest that alteration of the gut microbiota in patients with HE may be closely correlated to the levels of adipo-myokines in the blood.
Collapse
Affiliation(s)
- Bokyung Lee
- Department of Food Science and Nutrition, Dong A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
| | - Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - Jihyun Park
- Department of Health Sciences, Graduate School of Dong-A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
- Department of Health Sciences, Graduate School of Dong-A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Jeollanam-do, Republic of Korea
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
Samms RJ, Cheng CC, Fourcaudot M, Heikkinen S, Khattab A, Adams J, Cersosimo E, Triplitt C, Puckett C, Tsintzas K, Adams AC, Abdul-Ghani MA, DeFronzo RA, Norton L. FGF21 contributes to metabolic improvements elicited by combination therapy with exenatide and pioglitazone in patients with type 2 diabetes. Am J Physiol Endocrinol Metab 2022; 323:E123-E132. [PMID: 35723225 PMCID: PMC9291413 DOI: 10.1152/ajpendo.00050.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Fibroblast growth factor 21 (FGF21) is increased acutely by carbohydrate ingestion and is elevated in patients with type 2 diabetes (T2D). However, the physiological significance of increased FGF21 in humans remains largely unknown. We examined whether FGF21 contributed to the metabolic improvements observed following treatment of patients with T2D with either triple (metformin/pioglitazone/exenatide) or conventional (metformin/insulin/glipizide) therapy for 3 yr. Forty-six patients with T2D were randomized to receive either triple or conventional therapy to maintain HbA1c < 6.5%. A 2-h 75-g oral glucose tolerance test (OGTT) was performed at baseline and following 3 years of treatment to assess glucose tolerance, insulin sensitivity, and β-cell function. Plasma total and bioactive FGF21 levels were quantitated before and during the OGTT at both visits. Patients in both treatment arms experienced significant improvements in glucose control, but insulin sensitivity and β-cell function were markedly increased after triple therapy. At baseline, FGF21 levels were regulated acutely during the OGTT in both groups. After treatment, fasting total and bioactive FGF21 levels were significantly reduced in patients receiving triple therapy, but there was a relative increase in the proportion of bioactive FGF21 compared with that observed in conventionally treated subjects. Relative to baseline studies, triple therapy treatment also significantly modified FGF21 levels in response to a glucose load. These changes in circulating FGF21 were correlated with markers of improved glucose control and insulin sensitivity. Alterations in the plasma FGF21 profile may contribute to the beneficial metabolic effects of pioglitazone and exenatide in human patients with T2D.NEW & NOTEWORTHY In patients with T2D treated with a combination of metformin/pioglitazone/exenatide (triple therapy), we observed reduced total and bioactive plasma FGF21 levels and a relative increase in the proportion of circulating bioactive FGF21 compared with that in patients treated with metformin and sequential addition of glipizide and basal insulin glargine (conventional therapy). These data suggest that FGF21 may contribute, at least in part, to the glycemic benefits observed following combination therapy in patients with T2D.
Collapse
Affiliation(s)
| | | | - Marcel Fourcaudot
- Diabetes Division, University of Texas Health San Antonio, San Antonio, Texas
| | - Sami Heikkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Ahmed Khattab
- Diabetes Division, University of Texas Health San Antonio, San Antonio, Texas
| | - John Adams
- Diabetes Division, University of Texas Health San Antonio, San Antonio, Texas
| | - Eugenio Cersosimo
- Diabetes Division, University of Texas Health San Antonio, San Antonio, Texas
| | - Curtis Triplitt
- Diabetes Division, University of Texas Health San Antonio, San Antonio, Texas
| | - Curtis Puckett
- Diabetes Division, University of Texas Health San Antonio, San Antonio, Texas
| | - Kostas Tsintzas
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | - Luke Norton
- Diabetes Division, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
7
|
Haque N, Tischkau SA. Sexual Dimorphism in Adipose-Hypothalamic Crosstalk and the Contribution of Aryl Hydrocarbon Receptor to Regulate Energy Homeostasis. Int J Mol Sci 2022; 23:ijms23147679. [PMID: 35887027 PMCID: PMC9322714 DOI: 10.3390/ijms23147679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/16/2022] Open
Abstract
There are fundamental sex differences in the regulation of energy homeostasis. Better understanding of the underlying mechanisms of energy balance that account for this asymmetry will assist in developing sex-specific therapies for sexually dimorphic diseases such as obesity. Multiple organs, including the hypothalamus and adipose tissue, play vital roles in the regulation of energy homeostasis, which are regulated differently in males and females. Various neuronal populations, particularly within the hypothalamus, such as arcuate nucleus (ARC), can sense nutrient content of the body by the help of peripheral hormones such leptin, derived from adipocytes, to regulate energy homeostasis. This review summarizes how adipose tissue crosstalk with homeostatic network control systems in the brain, which includes energy regulatory regions and the hypothalamic–pituitary axis, contribute to energy regulation in a sex-specific manner. Moreover, development of obesity is contingent upon diet and environmental factors. Substances from diet and environmental contaminants can exert insidious effects on energy metabolism, acting peripherally through the aryl hydrocarbon receptor (AhR). Developmental AhR activation can impart permanent alterations of neuronal development that can manifest a number of sex-specific physiological changes, which sometimes become evident only in adulthood. AhR is currently being investigated as a potential target for treating obesity. The consensus is that impaired function of the receptor protects from obesity in mice. AhR also modulates sex steroid receptors, and hence, one of the objectives of this review is to explain why investigating sex differences while examining this receptor is crucial. Overall, this review summarizes sex differences in the regulation of energy homeostasis imparted by the adipose–hypothalamic axis and examines how this axis can be affected by xenobiotics that signal through AhR.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Shelley A. Tischkau
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Correspondence:
| |
Collapse
|
8
|
Pham HTA, Lee S, Lee YJ. Bicistronic reporter mice for monitoring of Fgf21 expression. Biochem Biophys Res Commun 2022; 619:104-109. [PMID: 35753217 DOI: 10.1016/j.bbrc.2022.06.045] [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: 05/26/2022] [Accepted: 06/14/2022] [Indexed: 11/02/2022]
Abstract
Fibroblast growth factor 21 (FGF21) is a metabolic hormone that is synthesized and secreted by cellular and metabolic stresses. Serum FGF21 levels are associated with clinical parameters in patients with various diseases, including metabolic disorders. Animal models that allow FGF21 levels to be monitored in vivo are important for research and clinical applications of FGF21. Here, a novel Fgf21-reporter mouse strain (Fgf21+/Luc2-tdT) expressing luciferase and tandem dimer tomato (tdT) fluorescence proteins under the control of the endogenous Fgf21 promoter was generated, which provided an in vitro and in vivo monitoring tool for the Fgf21 expression. Luciferase activity, in vivo bioluminescence, and tdT fluorescence were analyzed in adult mice fed or fasted for 24 h. Luciferase activities were significantly increased in the liver but slightly decreased in the pancreas of fasted mice compared with those of fed mice. In vivo bioluminescence signal was increased in the liver of fasted mice. Obvious tdT fluorescence was detected in the pancreas. These results suggest that Fgf21-reporter mice have great potential for research and clinical applications of FGF21.
Collapse
Affiliation(s)
- Huong Thi Anh Pham
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Gaetbeol-ro, Yeonsu-gu, Incheon, 21999, Republic of Korea.
| | - Sabin Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Gaetbeol-ro, Yeonsu-gu, Incheon, 21999, Republic of Korea.
| | - Young Jae Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Gaetbeol-ro, Yeonsu-gu, Incheon, 21999, Republic of Korea; Department of Biochemistry, College of Medicine, Gachon University, 155 Gaetbeol-ro, Yeonsu-gu, Incheon, 21999, Republic of Korea.
| |
Collapse
|
9
|
Wu YK, Ren ZN, Zhu SL, Wu YZ, Wang G, Zhang H, Chen W, He Z, Ye XL, Zhai QX. Sulforaphane ameliorates non-alcoholic fatty liver disease in mice by promoting FGF21/FGFR1 signaling pathway. Acta Pharmacol Sin 2022; 43:1473-1483. [PMID: 34654875 PMCID: PMC9159986 DOI: 10.1038/s41401-021-00786-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023]
Abstract
Most studies regarding the beneficial effect of sulforaphane (SFN) on non-alcoholic fatty liver disease (NAFLD) have focused on nuclear factor E2-related factor 2 (Nrf2). But the molecular mechanisms underlying the beneficial effect of SFN in the treatment of NAFLD remain controversial. Fibroblast growth factor (FGF) 21 is a member of the FGF family expressed mainly in liver but also in adipose tissue, muscle and pancreas, which functions as an endocrine factor and has been considered as a promising therapeutic candidate for the treatment of NAFLD. In the present study we investigated whether FGF21 was involved in the therapeutic effect of SFN against NAFLD. C57BL/6J mice were fed a high-fat diet (HFD) for 12 weeks to generate NAFLD and continued on the HFD for additional 6 weeks with or without SFN treatment. We showed that administration of SFN (0.56 g/kg) significantly ameliorated hepatic steatosis and inflammation in NAFLD mice, along with the improved glucose tolerance and insulin sensitivity, through suppressing the expression of proteins responsible for hepatic lipogenesis, while enhancing proteins for hepatic lipolysis and fatty acids oxidation. SFN administration significantly increased hepatic expression of FGFR1 and fibroblast growth factor 21 (FGF21) in NAFLD mice, along with decreased phosphorylation of p38 MAPK (the downstream of FGF21). HepG2 cells were treated in vitro with FFAs (palmitic acid and oleic acid) followed by different concentrations of SFN. We showed that the effects of SFN on FGF21 and FGFR1 protein expression were replicated in FFAs-treated HepG2 cells. Moreover, the increased FGFR1 protein occurred earlier than increased FGF21 protein. Interestingly, the rapid effect of SFN on FGFR1 protein was not regulated by the FGFR1 gene transcription. Knockdown of FGFR1 and p38 genes weakened SFN-reduced lipid deposition in FFAs-treated HepG2 cells. SFN administration in combination with rmFGF21 (1.5 mg/kg, i.p., every other day) for 3 weeks further suppressed hepatic steatosis in NAFLD mice. In conclusion, SFN ameliorates lipid metabolism disorders in NAFLD mice by upregulating FGF21/FGFR1 pathway. Our results verify that SFN may become a promising intervention to treat or relieve NAFLD.
Collapse
Affiliation(s)
- Yi-kuan Wu
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 China
| | - Zheng-nan Ren
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 China
| | - Sheng-long Zhu
- grid.258151.a0000 0001 0708 1323School of Medicine, Jiangnan University, Wuxi, 214122 China
| | - Yun-zhou Wu
- grid.412243.20000 0004 1760 1136College of Life Science, Northeast Agricultural University, Harbin, 150038 China
| | - Gang Wang
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 China
| | - Hao Zhang
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122 China
| | - Wei Chen
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122 China
| | - Zhao He
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021 China ,grid.27255.370000 0004 1761 1174School of Medicine, Shandong University, Jinan, 250012 China
| | - Xian-long Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000 China
| | - Qi-xiao Zhai
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122 China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 China
| |
Collapse
|
10
|
Szczepańska E, Gietka-Czernel M. FGF21: A Novel Regulator of Glucose and Lipid Metabolism and Whole-Body Energy Balance. Horm Metab Res 2022; 54:203-211. [PMID: 35413740 DOI: 10.1055/a-1778-4159] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Fibroblast growth factor (FGF) 21 is a recently recognized metabolic regulator that evokes interest due to its beneficial action of maintaining whole-body energy balance and protecting the liver from excessive triglyceride production and storage. Together with FGF19 and FGF23, FGF21 belongs to the FGF family with hormone-like activity. Serum FGF21 is generated primarily in the liver under nutritional stress stimuli like prolonged fasting or the lipotoxic diet, but also during increased mitochondrial and endoplasmic reticulum stress. FGF21 exerts its endocrine action in the central nervous system and adipose tissue. Acting in the ventromedial hypothalamus, FGF21 diminishes simple sugar intake. In adipose tissue, FGF21 promotes glucose utilization and increases energy expenditure by enhancing adipose tissue insulin sensitivity and brown adipose tissue thermogenesis. Therefore, FGF21 favors glucose consumption for heat production instead of energy storage. Furthermore, FGF21 specifically acts in the liver, where it protects hepatocytes from metabolic stress caused by lipid overload. FGF21 stimulates hepatic fatty acid oxidation and reduces lipid flux into the liver by increasing peripheral lipoprotein catabolism and reducing adipocyte lipolysis. Paradoxically, and despite its beneficial action, FGF21 is elevated in insulin resistance states, that is, fatty liver, obesity, and type 2 diabetes.
Collapse
Affiliation(s)
- Ewa Szczepańska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | | |
Collapse
|
11
|
Spann RA, Morrison CD, den Hartigh LJ. The Nuanced Metabolic Functions of Endogenous FGF21 Depend on the Nature of the Stimulus, Tissue Source, and Experimental Model. Front Endocrinol (Lausanne) 2022; 12:802541. [PMID: 35046901 PMCID: PMC8761941 DOI: 10.3389/fendo.2021.802541] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/09/2021] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a hormone that is involved in the regulation of lipid, glucose, and energy metabolism. Pharmacological FGF21 administration promotes weight loss and improves insulin sensitivity in rodents, non-human primates, and humans. However, pharmacologic effects of FGF21 likely differ from its physiological effects. Endogenous FGF21 is produced by many cell types, including hepatocytes, white and brown adipocytes, skeletal and cardiac myocytes, and pancreatic beta cells, and acts on a diverse array of effector tissues such as the brain, white and brown adipose tissue, heart, and skeletal muscle. Different receptor expression patterns dictate FGF21 function in these target tissues, with the primary effect to coordinate responses to nutritional stress. Moreover, different nutritional stimuli tend to promote FGF21 expression from different tissues; i.e., fasting induces hepatic-derived FGF21, while feeding promotes white adipocyte-derived FGF21. Target tissue effects of FGF21 also depend on its capacity to enter the systemic circulation, which varies widely from known FGF21 tissue sources in response to various stimuli. Due to its association with obesity and non-alcoholic fatty liver disease, the metabolic effects of endogenously produced FGF21 during the pathogenesis of these conditions are not well known. In this review, we will highlight what is known about endogenous tissue-specific FGF21 expression and organ cross-talk that dictate its diverse physiological functions, with particular attention given to FGF21 responses to nutritional stress. The importance of the particular experimental design, cellular and animal models, and nutritional status in deciphering the diverse metabolic functions of endogenous FGF21 cannot be overstated.
Collapse
Affiliation(s)
- Redin A. Spann
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Christopher D. Morrison
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Laura J. den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
12
|
Liao CY, Kummert OMP, Bair AM, Alavi N, Alavi J, Miller DM, Bagga I, Schempf AM, Hsu YM, Woods Ii BD, Brown Mayfield SM, Mitchell AN, Tannady G, Talbot AR, Dueck AM, Barrera Ovando R, Parker HD, Wang J, Schoeneweis JK, Kennedy BK. The autophagy inducer spermidine protects against metabolic dysfunction during overnutrition. J Gerontol A Biol Sci Med Sci 2021; 76:1714-1725. [PMID: 34060628 DOI: 10.1093/gerona/glab145] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Indexed: 12/16/2022] Open
Abstract
Autophagy, a process catabolizing intracellular components to maintain energy homeostasis, impacts aging and metabolism. Spermidine, a natural polyamine and autophagy activator, extends lifespan across a variety of species, including mice. In addition to protecting cardiac and liver tissue, spermidine also affects adipose tissue through unexplored mechanisms. Here, we examined spermidine in the links between autophagy and systemic metabolism. Consistently, daily injection of spermidine delivered even at late life is sufficient to cause a trend in lifespan extension in wild type mice. We further found that spermidine has minimal metabolic effects in young and old mice under normal nutrition. However, spermidine counteracts HFD (high-fat diet)-induced obesity by increasing lipolysis in visceral fat. Mechanistically, spermidine increases the hepatokine FGF21 expression in liver without reducing food intake. Spermidine also modulates FGF21 in adipose tissues, elevating FGF21 expression in subcutaneous fat, but reducing it in visceral fat. Despite this, FGF21 is not required for spermidine action, since Fgf21 -/- mice were still protected from HFD. Furthermore, the enhanced lipolysis by spermidine was also independent of autophagy in adipose tissue, given that adipose-specific autophagy deficient (Beclin-1 flox/+ Fabp4-cre) mice remained spermidine-responsive under HFD. Our results suggest that the metabolic effects of spermidine occurs through systemic changes in metabolism, involving multiple mechanistic pathways.
Collapse
Affiliation(s)
- Chen-Yu Liao
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Amanda M Bair
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Nora Alavi
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Josef Alavi
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Isha Bagga
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Yueh-Mei Hsu
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | | | | | | | | | - Aaron M Dueck
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | | | - Junying Wang
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Brian K Kennedy
- Buck Institute for Research on Aging, Novato, CA, USA.,Healthy Longevity Programme, Yong Loo Lin of Medicine, National University Singapore.,Centre for Healthy Longevity, National University Health System, Singapore.,Singapore Institute for Clinical Sciences, Singapore
| |
Collapse
|
13
|
Lu W, Li X, Luo Y. FGF21 in obesity and cancer: New insights. Cancer Lett 2020; 499:5-13. [PMID: 33264641 DOI: 10.1016/j.canlet.2020.11.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/23/2020] [Accepted: 11/23/2020] [Indexed: 02/08/2023]
Abstract
The endocrine FGF21 was discovered as a novel metabolic regulator in 2005 with new functions bifurcating from the canonic heparin-binding FGFs that directly promote cell proliferation and growth independent of a co-receptor. Early studies have demonstrated that FGF21 is a stress sensor in the liver and possibly, several other endocrine and metabolic tissues. Hepatic FGF21 signals via endocrine routes to quench episodes of metabolic derangements, promoting metabolic homeostasis. The convergence of mouse and human studies shows that FGF21 promotes lipid catabolism, including lipolysis, fatty acid oxidation, mitochondrial oxidative activity, and thermogenic energy dissipation, rather than directly regulating insulin and appetite. The white and brown adipose tissues and, to some extent, the hypothalamus, all of which host a transmembrane receptor binary complex of FGFR1 and co-receptor KLB, are considered the essential tissue and molecular targets of hepatic or pharmacological FGF21. On the other hand, a growing body of work has revealed that pancreatic acinar cells form a constitutive high-production site for FGF21, which then acts in an autocrine or paracrine mode. Beyond regulation of macronutrient metabolism and physiological energy expenditure, FGF21 appears to function in forestalling the development of fatty pancreas, steato-pancreatitis, fatty liver, and steato-hepatitis, thereby preventing the development of advanced pathologies such as pancreatic ductal adenocarcinoma or hepatocellular carcinoma. This review is intended to provide updates on these new discoveries that illuminate the protective roles of FGF21-FGFR1-KLB signal pathway in metabolic anomalies-associated severe tissue damage and malignancy, and to inform potential new preventive or therapeutic strategies for obesity-inflicted cancer patients via reducing metabolic risks and inflammation.
Collapse
Affiliation(s)
- Weiqin Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Yongde Luo
- Division of Gastroenterology and Hepatology, Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA; School of Pharmaceutical Science, Wenzhou Medical University, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Centeer BioTherapeutics Ltd Co, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Guo C, Zhao L, Li Y, Deng X, Yuan G. Relationship between FGF21 and drug or nondrug therapy of type 2 diabetes mellitus. J Cell Physiol 2020; 236:55-67. [PMID: 32583417 DOI: 10.1002/jcp.29879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 01/06/2023]
Abstract
Sedentary and high-calorie diets are associated with increased risk of obesity and type 2 diabetes mellitus, while exercise and diet control are also important nondrug treatments for diabetes. Fibroblast growth factor 21 (FGF21) is an important cytokine, which is mainly expressed in liver, fat and muscle tissue responding to nutrition and exercise, and plays an important role in the improvement of glucose and lipid metabolism. Due to the increasing serum FGF21 level in obesity and diabetes, FGF21 can be used as a predictor or biomarker of diabetes. A variety of clinical antidiabetic drugs can reduce the content of FGF21, possibly for the improvement of FGF21 sensitivity. In this paper, we reviewed the interactions between FGF21 and nondrug therapy (diet and exercise) for diabetes and explored the potential value of the combined application of clinical antidiabetic drugs and FGF21.
Collapse
Affiliation(s)
- Chang Guo
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Li Zhao
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yanyan Li
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xia Deng
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
15
|
Henriksson E, Andersen B. FGF19 and FGF21 for the Treatment of NASH-Two Sides of the Same Coin? Differential and Overlapping Effects of FGF19 and FGF21 From Mice to Human. Front Endocrinol (Lausanne) 2020; 11:601349. [PMID: 33414764 PMCID: PMC7783467 DOI: 10.3389/fendo.2020.601349] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
FGF19 and FGF21 analogues are currently in clinical development for the potential treatment of NASH. In Phase 2 clinical trials analogues of FGF19 and FGF21 decrease hepatic steatosis with up to 70% (MRI-PDFF) after 12 weeks and as early as 12-16 weeks of treatment an improvement in NASH resolution and fibrosis has been observed. Therefore, this class of compounds is currently of great interest in the field of NASH. FGF19 and FGF21 belong to the endocrine FGF19 subfamily and both require the co-receptor beta-klotho for binding and signalling through the FGF receptors. FGF19 is expressed in the ileal enterocytes and is released into the enterohepatic circulation in response to bile acids stimuli and in the liver FGF19 inhibits hepatic bile acids synthesis by transcriptional regulation of Cyp7A1, which is the rate limiting enzyme. FGF21 is, on the other hand, highly expressed in the liver and is released in response to high glucose, high free-fatty acids and low amino-acid supply and regulates energy, glucose and lipid homeostasis by actions in the CNS and in the adipose tissue. FGF19 and FGF21 are differentially expressed, have distinct target tissues and separate physiological functions. It is therefore of peculiar interest to understand why treatment with both FGF19 and FGF21 analogues have strong beneficial effects on NASH parameters in mice and human and whether the mode of action is overlapping This review will highlight the physiological and pharmacological effects of FGF19 and FGF21. The potential mode of action behind the anti-steatotic, anti-inflammatory and anti-fibrotic effects of FGF19 and FGF21 will be discussed. Finally, development of drugs is always a risk benefit analysis and the human relevance of adverse effects observed in pre-clinical species as well as findings in humans will be discussed. The aim is to provide a comprehensive overview of the current understanding of this drug class for the potential treatment of NASH.
Collapse
|
16
|
Luo Y, Yang Y, Liu M, Wang D, Wang F, Bi Y, Ji J, Li S, Liu Y, Chen R, Huang H, Wang X, Swidnicka-Siergiejko AK, Janowitz T, Beyaz S, Wang G, Xu S, Bialkowska AB, Luo CK, Pin CL, Liang G, Lu X, Wu M, Shroyer KR, Wolff RA, Plunkett W, Ji B, Li Z, Li E, Li X, Yang VW, Logsdon CD, Abbruzzese JL, Lu W. Oncogenic KRAS Reduces Expression of FGF21 in Acinar Cells to Promote Pancreatic Tumorigenesis in Mice on a High-Fat Diet. Gastroenterology 2019; 157:1413-1428.e11. [PMID: 31352001 PMCID: PMC6815712 DOI: 10.1053/j.gastro.2019.07.030] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 07/02/2019] [Accepted: 07/19/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND & AIMS Obesity is a risk factor for pancreatic cancer. In mice, a high-fat diet (HFD) and expression of oncogenic KRAS lead to development of invasive pancreatic ductal adenocarcinoma (PDAC) by unknown mechanisms. We investigated how oncogenic KRAS regulates the expression of fibroblast growth factor 21, FGF21, a metabolic regulator that prevents obesity, and the effects of recombinant human FGF21 (rhFGF21) on pancreatic tumorigenesis. METHODS We performed immunohistochemical analyses of FGF21 levels in human pancreatic tissue arrays, comprising 59 PDAC specimens and 45 nontumor tissues. We also studied mice with tamoxifen-inducible expression of oncogenic KRAS in acinar cells (KrasG12D/+ mice) and fElasCreERT mice (controls). KrasG12D/+ mice were placed on an HFD or regular chow diet (control) and given injections of rhFGF21 or vehicle; pancreata were collected and analyzed by histology, immunoblots, quantitative polymerase chain reaction, and immunohistochemistry. We measured markers of inflammation in the pancreas, liver, and adipose tissue. Activity of RAS was measured based on the amount of bound guanosine triphosphate. RESULTS Pancreatic tissues of mice expressed high levels of FGF21 compared with liver tissues. FGF21 and its receptor proteins were expressed by acinar cells. Acinar cells that expressed KrasG12D/+ had significantly lower expression of Fgf21 messenger RNA compared with acinar cells from control mice, partly due to down-regulation of PPARG expression-a transcription factor that activates Fgf21 transcription. Pancreata from KrasG12D/+ mice on a control diet and given injections of rhFGF21 had reduced pancreatic inflammation, infiltration by immune cells, and acinar-to-ductal metaplasia compared with mice given injections of vehicle. HFD-fed KrasG12D/+ mice given injections of vehicle accumulated abdominal fat, developed extensive inflammation, pancreatic cysts, and high-grade pancreatic intraepithelial neoplasias (PanINs); half the mice developed PDAC with liver metastases. HFD-fed KrasG12D/+ mice given injections of rhFGF21 had reduced accumulation of abdominal fat and pancreatic triglycerides, fewer pancreatic cysts, reduced systemic and pancreatic markers of inflammation, fewer PanINs, and longer survival-only approximately 12% of the mice developed PDACs, and none of the mice had metastases. Pancreata from HFD-fed KrasG12D/+ mice given injections of rhFGF21 had lower levels of active RAS than from mice given vehicle. CONCLUSIONS Normal acinar cells from mice and humans express high levels of FGF21. In mice, acinar expression of oncogenic KRAS significantly reduces FGF21 expression. When these mice are placed on an HFD, they develop extensive inflammation, pancreatic cysts, PanINs, and PDACs, which are reduced by injection of FGF21. FGF21 also reduces the guanosine triphosphate binding capacity of RAS. FGF21 might be used in the prevention or treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yongde Luo
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Medicine, Stony Brook University, Stony Brook, New York.
| | - Yaying Yang
- Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Muyun Liu
- Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dan Wang
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Feng Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yawei Bi
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Juntao Ji
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Suyun Li
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Yan Liu
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rong Chen
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Haojie Huang
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaojie Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Guoqiang Wang
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sulan Xu
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | | | - Catherine K. Luo
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Christoph L. Pin
- Departments of Pediatrics, Oncology, and Physiology and Pharmacology, Schulich School of Medicine, University of Western Ontario Children’s Health Research Institute, London, ON, Canana N5C 2V5
| | - Guang Liang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine. Indianapolis, IN, USA
| | - Maoxin Wu
- Department of Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Kenneth R. Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - William Plunkett
- Department of Experimental Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Baoan Ji
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Shanghai, China
| | - Ellen Li
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Vincent W. Yang
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Craig D. Logsdon
- Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA,Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - James L. Abbruzzese
- Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA,Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, NC, 27710, USA
| | - Weiqin Lu
- Department of Medicine, Stony Brook University, Stony Brook, New York; Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
17
|
Abstract
Members of the fibroblast growth factor (FGF) family play pleiotropic roles in cellular and metabolic homeostasis. During evolution, the ancestor FGF expands into multiple members by acquiring divergent structural elements that enable functional divergence and specification. Heparan sulfate-binding FGFs, which play critical roles in embryonic development and adult tissue remodeling homeostasis, adapt to an autocrine/paracrine mode of action to promote cell proliferation and population growth. By contrast, FGF19, 21, and 23 coevolve through losing binding affinity for extracellular matrix heparan sulfate while acquiring affinity for transmembrane α-Klotho (KL) or β-KL as a coreceptor, thereby adapting to an endocrine mode of action to drive interorgan crosstalk that regulates a broad spectrum of metabolic homeostasis. FGF19 metabolic axis from the ileum to liver negatively controls diurnal bile acid biosynthesis. FGF21 metabolic axes play multifaceted roles in controlling the homeostasis of lipid, glucose, and energy metabolism. FGF23 axes from the bone to kidney and parathyroid regulate metabolic homeostasis of phosphate, calcium, vitamin D, and parathyroid hormone that are important for bone health and systemic mineral balance. The significant divergence in structural elements and multiple functional specifications of FGF19, 21, and 23 in cellular and organismal metabolism instead of cell proliferation and growth sufficiently necessitate a new unified and specific term for these three endocrine FGFs. Thus, the term "FGF Metabolic Axis," which distinguishes the unique pathways and functions of endocrine FGFs from other autocrine/paracrine mitogenic FGFs, is coined.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
18
|
Pol CJ, Pollak NM, Jurczak MJ, Zacharia E, Karagiannides I, Kyriazis ID, Ntziachristos P, Scerbo DA, Brown BR, Aifantis I, Shulman GI, Goldberg IJ, Drosatos K. Cardiac myocyte KLF5 regulates body weight via alteration of cardiac FGF21. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2125-2137. [PMID: 31029826 PMCID: PMC6614009 DOI: 10.1016/j.bbadis.2019.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 01/22/2023]
Abstract
Cardiac metabolism affects systemic energetic balance. Previously, we showed that Krüppel-like factor (KLF)-5 regulates cardiomyocyte PPARα and fatty acid oxidation-related gene expression in diabetes. We surprisingly found that cardiomyocyte-specific KLF5 knockout mice (αMHC-KLF5-/-) have accelerated diet-induced obesity, associated with increased white adipose tissue (WAT). Alterations in cardiac expression of the mediator complex subunit 13 (Med13) modulates obesity. αMHC-KLF5-/- mice had reduced cardiac Med13 expression likely because KLF5 upregulates Med13 expression in cardiomyocytes. We then investigated potential mechanisms that mediate cross-talk between cardiomyocytes and WAT. High fat diet-fed αMHC-KLF5-/- mice had increased levels of cardiac and plasma FGF21, while food intake, activity, plasma leptin, and natriuretic peptides expression were unchanged. Consistent with studies reporting that FGF21 signaling in WAT decreases sumoylation-driven PPARγ inactivation, αMHC-KLF5-/- mice had less SUMO-PPARγ in WAT. Increased diet-induced obesity found in αMHC-KLF5-/- mice was absent in αMHC-[KLF5-/-;FGF21-/-] double knockout mice, as well as in αMHC-FGF21-/- mice that we generated. Thus, cardiomyocyte-derived FGF21 is a component of pro-adipogenic crosstalk between heart and WAT.
Collapse
Affiliation(s)
- Christine J Pol
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Nina M Pollak
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Michael J Jurczak
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Effimia Zacharia
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Iordanes Karagiannides
- Inflammatory Bowel Disease Center and Neuroendocrine Assay Core, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ioannis D Kyriazis
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Panagiotis Ntziachristos
- Howard Hughes Medical Institute, Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Diego A Scerbo
- Division of Preventive Medicine and Nutrition, Columbia University, New York, NY 10032, USA
| | - Brett R Brown
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA
| | - Iannis Aifantis
- Howard Hughes Medical Institute, Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Gerald I Shulman
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ira J Goldberg
- Division of Preventive Medicine and Nutrition, Columbia University, New York, NY 10032, USA
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Department of Pharmacology, Philadelphia, USA.
| |
Collapse
|
19
|
Fibroblast growth factor 8b induces uncoupling protein 1 expression in epididymal white preadipocytes. Sci Rep 2019; 9:8470. [PMID: 31186471 PMCID: PMC6560125 DOI: 10.1038/s41598-019-44878-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 05/20/2019] [Indexed: 02/08/2023] Open
Abstract
The number of brown adipocytes residing within murine white fat depots (brite adipocytes) varies a lot by depot, strain and physiological condition. Several endocrine fibroblast growth factors are implicated in the regulation of brite adipocyte abundance. The family of fibroblast growth factors can be categorized by their site of action into endocrine, paracrine and intracellular peptides. We here screened paracrine fibroblast growth factors for their potential to drive brite adipogenesis in differentiating epididymal white adipocytes and identified fibroblast growth factor 8b to induce uncoupling protein 1 expression, but at the same time to interfere in adipogenesis. In an in vivo trial, fibroblast growth factor 8b released into the epididymal fat depot failed to robustly increase the number of brite adipocytes. The specific action of fibroblast growth factor 8b on the uncoupling protein 1 promoter in cultured epididymal adipocytes provides a model system to dissect specific gene regulatory networks.
Collapse
|
20
|
Broekema M, Savage D, Monajemi H, Kalkhoven E. Gene-gene and gene-environment interactions in lipodystrophy: Lessons learned from natural PPARγ mutants. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:715-732. [PMID: 30742913 DOI: 10.1016/j.bbalip.2019.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/13/2019] [Accepted: 02/02/2019] [Indexed: 12/13/2022]
|
21
|
Lee KW, Jeong JY, An YJ, Lee JH, Yim HS. FGF11 influences 3T3-L1 preadipocyte differentiation by modulating the expression of PPARγ regulators. FEBS Open Bio 2019; 9:769-780. [PMID: 30984550 PMCID: PMC6443871 DOI: 10.1002/2211-5463.12619] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/31/2019] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor 11 (FGF11) is a member of the intracellular fibroblast growth factor superfamily. Here, we identified FGF11 as a novel mediator of adipogenesis. During 3T3‐L1 adipocyte differentiation, the expression of FGF11 decreased at the mitotic clonal expansion stage and increased at the terminal differentiation stage. FGF11 knockdown reduced the expression of peroxisome proliferator‐activated receptor gamma (PPARγ), a master regulator of adipogenesis, resulting in the inhibition of adipocyte differentiation. Treatment with the PPARγ agonist rosiglitazone restored the inhibition of adipogenesis caused by FGF11 knockdown. We also report that the expression of the PPARγ regulators CCAAT/enhancer‐binding protein α, sterol regulatory element‐binding protein 1, KLF9, KLF2, GATA binding factor 2, and GATA binding factor 3 was influenced by FGF11. These results suggest that FGF11 indirectly controls the expression of PPARγ through modifying the expression of multiple PPARγ regulators, thereby mediating adipogenesis.
Collapse
Affiliation(s)
- Kyeong Won Lee
- Marine Biotechnology Research Center Korea Institute of Ocean Science and Technology Busan Korea
| | - Jae-Yeon Jeong
- Marine Biotechnology Research Center Korea Institute of Ocean Science and Technology Busan Korea
| | - Young Jun An
- Marine Biotechnology Research Center Korea Institute of Ocean Science and Technology Busan Korea
| | - Jung-Hyun Lee
- Marine Biotechnology Research Center Korea Institute of Ocean Science and Technology Busan Korea.,Department of Marine Biotechnology Korea University of Science and Technology Daejeon Korea
| | - Hyung-Soon Yim
- Marine Biotechnology Research Center Korea Institute of Ocean Science and Technology Busan Korea
| |
Collapse
|
22
|
Cuevas-Ramos D, Mehta R, Aguilar-Salinas CA. Fibroblast Growth Factor 21 and Browning of White Adipose Tissue. Front Physiol 2019; 10:37. [PMID: 30804796 PMCID: PMC6370737 DOI: 10.3389/fphys.2019.00037] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/14/2019] [Indexed: 12/30/2022] Open
Abstract
Interest has been focused on differentiating anatomical, molecular, and physiological characteristics of the types of mammalian adipose tissues. White adipose tissue (WAT) and brown adipose tissue (BAT) are the two main forms of adipose tissue in humans. WAT functions as an endocrine organ and serves as a reservoir of energy in the form of triglycerides. The hormones released by WAT are called adipokines. BAT consists of a group of specialized cells with abundant uncoupling protein 1 (UCP1) in the inner mitochondrial membrane and also fulfills endocrine functions. Following the identification of functional (BAT) in human adults, there has been a great deal of interest in finding out how it is induced, its localization, and the mechanisms by which it regulates thermogenesis. Fibroblast growth factor 21 (FGF21) is a key regulator of the differentiation to brown adipocytes. The main mechanisms occur through enhancing UCP1 expression. In addition, following exposure to cold or exercise, FGF21 induces upregulation of local peroxisome proliferator-activated receptor gamma co-activator (PGC)-1-alfa and thus promotes thermogenesis in adipose tissue and skeletal muscle. FGF21 integrates several pathways allowing the regulation of human energy balance, glucose levels, and lipid metabolism. Such mechanisms and their clinical relevance are summarized in this review.
Collapse
Affiliation(s)
- Daniel Cuevas-Ramos
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - R Mehta
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carlos A Aguilar-Salinas
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Instituto Tecnológico y de Estudios Superiores de Monterrey Tec Salud, Monterrey, Mexico
| |
Collapse
|
23
|
Cai G, Liu J, Wang M, Su L, Cai M, Huang K, Li X, Li M, Wang L, Huang X. Mutual promotion of FGF21 and PPARγ attenuates hypoxia-induced pulmonary hypertension. Exp Biol Med (Maywood) 2019; 244:252-261. [PMID: 30714402 DOI: 10.1177/1535370219828692] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
IMPACT STATEMENT In this study, we reported for the first time that FGF21 alleviated hypoxia-induced pulmonary hypertension through attenuation of increased pulmonary arterial pressure, pulmonary arterial remodeling and collagen deposition in vivo, and we confirmed the mutual promotion of FGF21 and PPARγ in hypoxia-induced pulmonary hypertension. Additionally, we found that FGF21 and PPARγ mutually promote each other's expression via the AMPK/PGC-1α pathway and KLB protein in vitro and in vivo. Pulmonary hypertension is a progressive and serious pathological phenomenon with a poor prognosis, and current therapies are highly limited. Our results provide novel insight into potential clinical therapies for pulmonary hypertension and establish the possibility of using this drug combination and potential dosage reductions in clinical settings.
Collapse
Affiliation(s)
- Gexiang Cai
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Jingjing Liu
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Meibin Wang
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Lihuang Su
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Mengsi Cai
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Kate Huang
- 2 Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, PR China
| | - Xiuchun Li
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Manxiang Li
- 3 Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, PR China
| | - Liangxing Wang
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Xiaoying Huang
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| |
Collapse
|
24
|
Ferrer‐Curriu G, Redondo‐Angulo I, Guitart‐Mampel M, Ruperez C, Mas‐Stachurska A, Sitges M, Garrabou G, Villarroya F, Fernández‐Solà J, Planavila A. Fibroblast growth factor‐21 protects against fibrosis in hypertensive heart disease. J Pathol 2019; 248:30-40. [DOI: 10.1002/path.5226] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/16/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Gemma Ferrer‐Curriu
- Departament de Bioquímica i Biologia MolecularInstitut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona Barcelona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Barcelona Spain
| | - Ibon Redondo‐Angulo
- Departament de Bioquímica i Biologia MolecularInstitut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona Barcelona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Barcelona Spain
| | - Mariona Guitart‐Mampel
- Muscle Research and Mitochondrial Function LaboratoryCellex‐IDIBAPS, Faculty of Medicine and Health Science, University of Barcelona, Internal Medicine Service, Hospital Clínic of Barcelona, Barcelona and CIBERER Barcelona Spain
| | - Celia Ruperez
- Departament de Bioquímica i Biologia MolecularInstitut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona Barcelona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Barcelona Spain
| | - Aleksandra Mas‐Stachurska
- Cardiology DepartmentThorax Institute, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona Barcelona Spain
| | - Marta Sitges
- Cardiology DepartmentThorax Institute, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona Barcelona Spain
| | - Gloria Garrabou
- Muscle Research and Mitochondrial Function LaboratoryCellex‐IDIBAPS, Faculty of Medicine and Health Science, University of Barcelona, Internal Medicine Service, Hospital Clínic of Barcelona, Barcelona and CIBERER Barcelona Spain
| | - Francesc Villarroya
- Departament de Bioquímica i Biologia MolecularInstitut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona Barcelona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Barcelona Spain
| | | | - Anna Planavila
- Departament de Bioquímica i Biologia MolecularInstitut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona Barcelona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) Barcelona Spain
| |
Collapse
|
25
|
Abstract
The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is a master regulator of adipocyte differentiation and is the target for the insulin-sensitizing thiazolidinedione (TZD) drugs used to treat type 2 diabetes. In cell-based in vitro studies, the transcriptional activity of PPARγ is inhibited by covalent attachment of small ubiquitin-related modifier (SUMOylation) at K107 in its N terminus. However, whether this posttranslational modification is relevant in vivo remains unclear. Here, using mice homozygous for a mutation (K107R) that prevents SUMOylation at this position, we demonstrate that PPARγ is SUMOylated at K107 in white adipose tissue. We further show that in the context of diet-induced obesity PPARγ-K107R-mutant mice have enhanced insulin sensitivity without the corresponding increase in adiposity that typically accompanies PPARγ activation by TZDs. Accordingly, the PPARγ-K107R mutation was weaker than TZD treatment in stimulating adipocyte differentiation in vitro. Moreover, we found that both the basal and TZD-dependent transcriptomes of inguinal and epididymal white adipose tissue depots were markedly altered in the K107R-mutant mice. We conclude that PPARγ SUMOylation at K107 is physiologically relevant and may serve as a pharmacologic target for uncoupling PPARγ's beneficial insulin-sensitizing effect from its adverse effect of weight gain.
Collapse
|
26
|
Yan X, Gou Z, Li Y, Wang Y, Zhu J, Xu G, Zhang Q. Fibroblast growth factor 21 inhibits atherosclerosis in apoE-/- mice by ameliorating Fas-mediated apoptosis. Lipids Health Dis 2018; 17:203. [PMID: 30157856 PMCID: PMC6114502 DOI: 10.1186/s12944-018-0846-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/07/2018] [Indexed: 12/02/2022] Open
Abstract
Background FGF21 is a critical endogenous regulator in energy homeostasis and systemic glucose and lipid metabolism. Despite intensive study of the metabolic functions of FGF21, its important role in heart disease needs further exploration. Apoptosis induced by ox-LDL in vascular endothelial cells is an important step in the progress of atherosclerosis. Methods The effects of FGF21 treatment on apoptosis induced by ox-LDL were tested in HUVECs. The role of FGF21 in atherosclerosis was studied by evaluating its function in apolipoprotein E double knockout (apoE−/−) mice. Results We found that apoptosis in HUVECs was alleviated by FGF21 treatment. The effects of FGF21 were independent of the ERK1/2 pathway and were mediated through inhibition of the Fas signaling pathway. FGF21 suppressed the development of atherosclerosis, and the administration of FGF21 ameliorated Fas-mediated apoptosis in apoE−/− mice. Conclusion FGF21 protects against apoptosis in HUVECs by suppressing the expression of Fas; furthermore, FGF21 alleviated atherosclerosis by ameliorating Fas-mediated apoptosis in apoE−/− mice. Electronic supplementary material The online version of this article (10.1186/s12944-018-0846-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinxin Yan
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, People's Republic of China
| | - Zhongshan Gou
- Center for Medical Ultrasound, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, People's Republic of China
| | - Yuan Li
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, People's Republic of China
| | - Yu Wang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, People's Republic of China
| | - Jingyan Zhu
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, People's Republic of China
| | - Guidong Xu
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, People's Republic of China
| | - Qian Zhang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, People's Republic of China.
| |
Collapse
|
27
|
You D, Nilsson E, Tenen DE, Lyubetskaya A, Lo JC, Jiang R, Deng J, Dawes BA, Vaag A, Ling C, Rosen ED, Kang S. Dnmt3a is an epigenetic mediator of adipose insulin resistance. eLife 2017; 6:30766. [PMID: 29091029 PMCID: PMC5730374 DOI: 10.7554/elife.30766] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/29/2017] [Indexed: 12/21/2022] Open
Abstract
Insulin resistance results from an intricate interaction between genetic make-up and environment, and thus may be orchestrated by epigenetic mechanisms like DNA methylation. Here, we demonstrate that DNA methyltransferase 3a (Dnmt3a) is both necessary and sufficient to mediate insulin resistance in cultured mouse and human adipocytes. Furthermore, adipose-specific Dnmt3a knock-out mice are protected from diet-induced insulin resistance and glucose intolerance without accompanying changes in adiposity. Unbiased gene profiling studies revealed Fgf21 as a key negatively regulated Dnmt3a target gene in adipocytes with concordant changes in DNA methylation at the Fgf21 promoter region. Consistent with this, Fgf21 can rescue Dnmt3a-mediated insulin resistance, and DNA methylation at the FGF21 locus was elevated in human subjects with diabetes and correlated negatively with expression of FGF21 in human adipose tissue. Taken together, our data demonstrate that adipose Dnmt3a is a novel epigenetic mediator of insulin resistance in vitro and in vivo.
Collapse
Affiliation(s)
- Dongjoo You
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| | - Emma Nilsson
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Danielle E Tenen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, United States
| | | | - James C Lo
- Weill Cornell Medical College, New York, United States
| | - Rencong Jiang
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| | - Jasmine Deng
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| | - Brian A Dawes
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, United States
| | - Allan Vaag
- Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Early Clinical Development, AstraZeneca, Innovative Medicines, Göteborg, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Evan D Rosen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, United States.,Broad Institute of Harvard and MIT, Cambridge, United States
| | - Sona Kang
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
28
|
Staiger H, Keuper M, Berti L, Hrabe de Angelis M, Häring HU. Fibroblast Growth Factor 21-Metabolic Role in Mice and Men. Endocr Rev 2017; 38:468-488. [PMID: 28938407 DOI: 10.1210/er.2017-00016] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/25/2017] [Indexed: 12/18/2022]
Abstract
Since its identification in 2000, the interest of scientists in the hepatokine fibroblast growth factor (FGF) 21 has tremendously grown, and still remains high, due to a wealth of very robust data documenting this factor's favorable effects on glucose and lipid metabolism in mice. For more than ten years now, intense in vivo and ex vivo experimentation addressed the physiological functions of FGF21 in humans as well as its pathophysiological role and pharmacological effects in human metabolic disease. This work produced a comprehensive collection of data revealing overlaps in FGF21 expression and function but also significant differences between mice and humans that have to be considered before translation from bench to bedside can be successful. This review summarizes what is known about FGF21 in mice and humans with a special focus on this factor's role in glucose and lipid metabolism and in metabolic diseases, such as obesity and type 2 diabetes mellitus. We highlight the discrepancies between mice and humans and try to decipher their underlying reasons.
Collapse
Affiliation(s)
- Harald Staiger
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Interfaculty Center for Pharmacogenomics and Pharma Research, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Michaela Keuper
- Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Lucia Berti
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany.,Chair for Experimental Genetics, Technical University Munich, 85764 Neuherberg, Germany
| | - Hans-Ulrich Häring
- Interfaculty Center for Pharmacogenomics and Pharma Research, Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany.,German Center for Diabetes Research, 85764 Neuherberg, Germany.,Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
29
|
Huang Z, Zhong L, Lee JTH, Zhang J, Wu D, Geng L, Wang Y, Wong CM, Xu A. The FGF21-CCL11 Axis Mediates Beiging of White Adipose Tissues by Coupling Sympathetic Nervous System to Type 2 Immunity. Cell Metab 2017; 26:493-508.e4. [PMID: 28844880 DOI: 10.1016/j.cmet.2017.08.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/24/2017] [Accepted: 08/01/2017] [Indexed: 02/08/2023]
Abstract
Type 2 cytokines are important signals triggering biogenesis of thermogenic beige adipocytes in white adipose tissue (WAT) during cold acclimation. However, how cold activates type 2 immunity in WAT remains obscure. Here we show that cold-induced type 2 immune responses and beiging in subcutaneous WAT (scWAT) are abrogated in mice with adipose-selective ablation of FGF21 or its co-receptor β-Klotho, whereas such impairments are reversed by replenishment with chemokine CCL11. Mechanistically, FGF21 acts on adipocytes in an autocrine manner to promote the expression and secretion of CCL11 via activation of ERK1/2, which drives recruitment of eosinophils into scWAT, leading to increases in accumulation of M2 macrophages, and proliferation and commitment of adipocyte precursors into beige adipocytes. These FGF21-elicited type 2 immune responses and beiging are blocked by CCL11 neutralization. Thus, the adipose-derived FGF21-CCL11 axis triggers cold-induced beiging and thermogenesis by coupling sympathetic nervous system to activation of type 2 immunity in scWAT.
Collapse
Affiliation(s)
- Zhe Huang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ling Zhong
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jimmy Tsz Hang Lee
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jialiang Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Donghai Wu
- The Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Leiluo Geng
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Chi-Ming Wong
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, 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.
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, 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
|
30
|
Luo Y, Ye S, Chen X, Gong F, Lu W, Li X. Rush to the fire: FGF21 extinguishes metabolic stress, metaflammation and tissue damage. Cytokine Growth Factor Rev 2017; 38:59-65. [PMID: 28887067 DOI: 10.1016/j.cytogfr.2017.08.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 08/17/2017] [Indexed: 11/26/2022]
Abstract
FGF21 is a master regulator of homeostasis of local and systemic lipid, glucose and energy metabolism. Since its discovery a decade ago, significant progress has been made in understanding the basic molecular, cellular and physiological mechanisms underlying its metabolic roles, and characterizing its beneficial pharmacological activities and possible pathological roles in obesity, diabetes, dyslipidemia, fatty liver disease and their collateral complications and tissue damage. Under basal or normal conditions, FGF21 appears to play a dispensable role in metabolism. However, in response to a variety of cellular and metabolic stress, FGF21 is significantly upregulated to serve as a potent catabolic factor leading to the clearance of excessive lipids and glucose, and therefore, antagonizes metabolic and energy imbalance in a negative fashion. Furthermore, FGF21 treatment ameliorates tissue damage resulted from the harmful effects of metabolic abnormalities, which often ensue an oxidative, pro-inflammatory, inflammatory and/or immune stress state, the so-called metaflammation. Most notably, studies focusing on the liver, pancreas, cardio-vasculature and kidney have revealed its significant protective effects against the structural and functional damages induced by the obese, diabetic or other abnormal metabolic conditions. In this review, we will summarize the current progress on the roles of FGF21 against metaflammation and metabolic tissue damage.
Collapse
Affiliation(s)
- Yongde Luo
- School of Pharmaceutical Science, Wenzhou Medical University; Center for Collaborative Translational Biomedical Research, Wenzhou University, Wenzhou, Zhejiang 325000, China; Proteomics and Nanotechnology Laboratory, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030. Current address: Centeer BioTherapeutics Ltd Co., Houston, TX 77021.
| | - Sheng Ye
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiong Chen
- School of Pharmaceutical Science, Wenzhou Medical University; Center for Collaborative Translational Biomedical Research, Wenzhou University, Wenzhou, Zhejiang 325000, China
| | - Fanghua Gong
- School of Pharmaceutical Science, Wenzhou Medical University; Center for Collaborative Translational Biomedical Research, Wenzhou University, Wenzhou, Zhejiang 325000, China
| | - Weiqin Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University; Center for Collaborative Translational Biomedical Research, Wenzhou University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
31
|
Ong KL, O'Connell R, Januszewski AS, Jenkins AJ, Xu A, Sullivan DR, Barter PJ, Scott RS, Taskinen MR, Waldman B, Colman PG, Best JD, Simes JR, Rye KA, Keech AC. Baseline Circulating FGF21 Concentrations and Increase after Fenofibrate Treatment Predict More Rapid Glycemic Progression in Type 2 Diabetes: Results from the FIELD Study. Clin Chem 2017; 63:1261-1270. [DOI: 10.1373/clinchem.2016.270876] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/05/2017] [Indexed: 11/06/2022]
Abstract
Abstract
BACKGROUND
It is not known whether circulating fibroblast growth factor 21 (FGF21) concentrations are associated with glycemic progression in patients with established type 2 diabetes. This study reports this relationship in type 2 diabetes patients participating in the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) trial.
METHODS
Plasma FGF21 was quantified in 9697 study participants. Among patients with lifestyle-only glucose control measures at baseline, glycemic progression was defined as the initiation of oral hypoglycemic agents or insulin therapy. We assessed the relationship of FGF21 concentrations with glycohemoglobin (Hb A1c), the homeostasis model assessment of β-cell function (HOMA-B) and insulin resistance (HOMA-IR), and glycemic progression.
RESULTS
Among 2584 patients with lifestyle-only glycemic therapy at baseline, plasma FGF21 concentrations were positively associated with HOMA-IR (5.1% increase per 100% increase in FGF21 concentrations). Patients with higher baseline plasma FGF21 concentrations had higher risk of glycemic progression over a 5-year period (P = 0.02), but the association was not significant after further adjusting for alanine aminotransferase (ALT) enzyme activity. During the fenofibrate active run-in phase, higher tertiles of fenofibrate-induced increase in FGF21 concentrations were associated with higher risk of glycemic progression (adjusted hazards ratio = 1.09 and 1.18 for tertiles 2 and 3, respectively, P for trend = 0.01), even after adjusting for ALT enzyme activity. This association was statistically significant in the fenofibrate group only (P = 0.01).
CONCLUSIONS
Higher baseline and fenofibrate-induced increase in FGF21 concentrations predict more rapid glycemic progression in type 2 diabetes patients. This association may be partly explained by hepatic function.
Collapse
Affiliation(s)
- Kwok-Leung Ong
- Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Rachel O'Connell
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | | | - Alicia J Jenkins
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Aimin Xu
- Department of Medicine, University of Hong Kong, Hong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong
| | - David R Sullivan
- Department of Clinical Biochemistry, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Philip J Barter
- Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Russell S Scott
- Lipid and Diabetes Research Group, Christchurch Hospital, Christchurch, New Zealand
| | - Marja-Riitta Taskinen
- Heart and Lung Centre, Cardiovascular Research Unit, Helsinki University Central Hospital, Helsinki, Finland
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
| | - Boris Waldman
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Peter G Colman
- Department of Diabetes & Endocrinology, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - James D Best
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - John R Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Anthony C Keech
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
32
|
Li X, Stanislaus S, Asuncion F, Niu QT, Chinookoswong N, Villasenor K, Wang J, Wong P, Boyce R, Dwyer D, Han CY, Chen MM, Liu B, Stolina M, Ke HZ, Ominsky MS, Véniant MM, Xu J. FGF21 Is Not a Major Mediator for Bone Homeostasis or Metabolic Actions of PPARα and PPARγ Agonists. J Bone Miner Res 2017; 32:834-845. [PMID: 27505721 DOI: 10.1002/jbmr.2936] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 07/28/2016] [Accepted: 08/08/2016] [Indexed: 11/10/2022]
Abstract
Results of prior studies suggest that fibroblast growth factor 21 (FGF21) may be involved in bone turnover and in the actions of peroxisome proliferator-activated receptor (PPAR) α and γ in mice. We have conducted independent studies to examine the effects of FGF21 on bone homeostasis and the role of FGF21 in PPARα and γ actions. High-fat-diet-induced obesity (DIO) mice were administered vehicle or recombinant human FGF21 (rhFGF21) intraperitoneally at 0 (vehicle), 0.1, 1, and 3 mg/kg daily for 2 weeks. Additional groups of DIO mice received water or 10 mg/kg rosiglitazone daily. Mice treated with rhFGF21 or rosiglitazone showed expected metabolic improvements in glucose, insulin, and lipid levels. However, bone loss was not detected in rhFGF21-treated mice by dual-energy X-ray absorptiometry (DXA), micro-CT, and histomorphometric analyses. Mineral apposition rate, a key bone formation parameter, was unchanged by rhFGF21, while significantly decreased by rosiglitazone in DIO mice. Bone resorption markers, OPG/RANKL mRNA expression, and histological bone resorption indices were unchanged by rhFGF21 or rosiglitazone. Bone marrow fat was unchanged by rhFGF21, while increased by rosiglitazone. Furthermore, FGF21 knockout mice did not show high bone mass phenotype. Treatment with PPARα or PPARγ agonists caused similar metabolic effects in FGF21 knockout and wild-type mice. These results contrast with previous findings and suggest that FGF21 is not critical for bone homeostasis or actions of PPARα and PPARγ. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Shanaka Stanislaus
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Frank Asuncion
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Qing-Tian Niu
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Kelly Villasenor
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Jin Wang
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, CA, USA
| | - Philip Wong
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, CA, USA
| | - Rogely Boyce
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - Denise Dwyer
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Chun-Ya Han
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Michelle M Chen
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Benxian Liu
- Department of Inflammation, Amgen Inc., Thousand Oaks, CA, USA
| | - Marina Stolina
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Hua Zhu Ke
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Michael S Ominsky
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Murielle M Véniant
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Jing Xu
- Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| |
Collapse
|
33
|
Identification of Sp1 as a Transcription Activator to Regulate Fibroblast Growth Factor 21 Gene Expression. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8402035. [PMID: 28466020 PMCID: PMC5390607 DOI: 10.1155/2017/8402035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/08/2017] [Accepted: 03/16/2017] [Indexed: 12/04/2022]
Abstract
Fibroblast growth factor 21 (FGF21) is a metabolic hormone with multiple beneficial effects on lipid and glucose homeostasis. Previous study demonstrated that FGF21 might be one of the Sp1 target genes. However, the transcriptional role of Sp1 on FGF21 in adipose tissue and liver has not been reported. In this study, we found that the proximal promoter of mouse FGF21 is located between −63 and −20 containing two putative Sp1-binding sites. Sp1 is a mammalian transcription factor involved in the regulation of many genes during physiological and pathological processes. Our study showed that overexpression of Sp1 or suppressing Sp1 expression resulted in increased or reduced FGF21 promoter activity, respectively. Mutation analysis demonstrated that the Sp1-binding site located between −46 and −38 plays a primary role in transcription of FGF21. Electrophoretic mobility shift assay and chromatin immunoprecipitation analysis indicated that Sp1 specifically bound to this region. Furthermore, the binding activity of Sp1 was significantly increased in adipose tissues of HFD-induced obese mouse and liver of DEN-treated mouse. Thus, our results demonstrate that Sp1 positively regulates the basal transcription of FGF21 in the liver and adipose tissue and contributes to the obesity-induced FGF21 upregulation in mouse adipose tissue and hepatic FGF21 upregulation in hepatocarcinogenesis.
Collapse
|
34
|
Kharitonenkov A, DiMarchi R. Fibroblast growth factor 21 night watch: advances and uncertainties in the field. J Intern Med 2017; 281:233-246. [PMID: 27878865 DOI: 10.1111/joim.12580] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fibroblast growth factor (FGF) 21 belongs to a hormone-like subgroup within the FGF superfamily. The members of this subfamily, FGF19, FGF21 and FGF23, are characterized by their reduced binding affinity for heparin that enables them to be transported in the circulation and function in an endocrine manner. It is likely that FGF21 also acts in an autocrine and paracrine fashion, as multiple organs can produce this protein and its plasma concentration seems to be below the level necessary to induce a pharmacological effect. FGF21 signals via FGF receptors, but for efficient receptor engagement it requires a cofactor, membrane-spanning βKlotho (KLB). The regulation of glucose uptake in adipocytes was the initial biological activity ascribed to FGF21, but this hormone is now recognized to stimulate many other pathways in vitro and display multiple pharmacological effects in metabolically compromised animals and humans. Understanding of the precise physiology of FGF21 and its potential medicinal role has evolved exponentially over the last decade, yet numerous aspects remain to be defined and others are a source of debate. Here we provide a historical overview of the advances in FGF21 biology focusing on the uncertainties in the mechanism of action as well as the differing viewpoints relating to this intriguing protein.
Collapse
Affiliation(s)
- A Kharitonenkov
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, USA
| | - R DiMarchi
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, USA
| |
Collapse
|
35
|
Coate KC, Hernandez G, Thorne CA, Sun S, Le TDV, Vale K, Kliewer SA, Mangelsdorf DJ. FGF21 Is an Exocrine Pancreas Secretagogue. Cell Metab 2017; 25:472-480. [PMID: 28089565 PMCID: PMC5299054 DOI: 10.1016/j.cmet.2016.12.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 10/28/2016] [Accepted: 12/10/2016] [Indexed: 12/21/2022]
Abstract
The metabolic stress hormone FGF21 is highly expressed in exocrine pancreas, where its levels are increased by refeeding and chemically induced pancreatitis. However, its function in the exocrine pancreas remains unknown. Here, we show that FGF21 stimulates digestive enzyme secretion from pancreatic acinar cells through an autocrine/paracrine mechanism that requires signaling through a tyrosine kinase receptor complex composed of an FGF receptor and β-Klotho. Mice lacking FGF21 accumulate zymogen granules and are susceptible to pancreatic ER stress, an effect that is reversed by administration of recombinant FGF21. Mice carrying an acinar cell-specific deletion of β-Klotho also accumulate zymogen granules but are refractory to FGF21-stimulated secretion. Like the classical post-prandial secretagogue, cholecystokinin (CCK), FGF21 triggers intracellular calcium release via PLC-IP3R signaling. However, unlike CCK, FGF21 does not induce protein synthesis, thereby preventing protein accumulation. Thus, pancreatic FGF21 is a digestive enzyme secretagogue whose physiologic function is to maintain acinar cell proteostasis.
Collapse
Affiliation(s)
- Katie C Coate
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Genaro Hernandez
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Curtis A Thorne
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shengyi Sun
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thao D V Le
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kevin Vale
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Steven A Kliewer
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - David J Mangelsdorf
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
36
|
Abstract
Adipose tissue plays a central role in regulating whole-body energy and glucose homeostasis through its subtle functions at both organ and systemic levels. On one hand, adipose tissue stores energy in the form of lipid and controls the lipid mobilization and distribution in the body. On the other hand, adipose tissue acts as an endocrine organ and produces numerous bioactive factors such as adipokines that communicate with other organs and modulate a range of metabolic pathways. Moreover, brown and beige adipose tissue burn lipid by dissipating energy in the form of heat to maintain euthermia, and have been considered as a new way to counteract obesity. Therefore, adipose tissue dysfunction plays a prominent role in the development of obesity and its related disorders such as insulin resistance, cardiovascular disease, diabetes, depression and cancer. In this review, we will summarize the recent findings of adipose tissue in the control of metabolism, focusing on its endocrine and thermogenic function.
Collapse
Affiliation(s)
- Liping Luo
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Meilian Liu
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular BiologyUniversity of New Mexico Health Sciences Center,
Albuquerque, New Mexico, USA
| |
Collapse
|
37
|
Lundsgaard AM, Fritzen AM, Sjøberg KA, Myrmel LS, Madsen L, Wojtaszewski JFP, Richter EA, Kiens B. Circulating FGF21 in humans is potently induced by short term overfeeding of carbohydrates. Mol Metab 2016; 6:22-29. [PMID: 28123934 PMCID: PMC5220397 DOI: 10.1016/j.molmet.2016.11.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 01/05/2023] Open
Abstract
Objective Fibroblast-growth factor 21 (FGF21) is thought to be important in metabolic regulation. Recently, low protein diets have been shown to increase circulating FGF21 levels. However, when energy contribution from dietary protein is lowered, other macronutrients, such as carbohydrates, must be increased to meet eucaloric balance. This raises the possibility that intake of a diet rich in carbohydrates may induce an increase in plasma FGF21 levels per se. Here we studied the role of dietary carbohydrates on the levels of circulating FGF21 and concomitant physiologic effects by feeding healthy men a carbohydrate rich diet without reducing protein intake. Methods A diet enriched in carbohydrates (80 E% carbohydrate; CHO) and a eucaloric control diet (CON) were provided to nine healthy men for three days. The energy intake during the CHO diet was increased (+75% energy) to ensure similar dietary protein intake in CHO and CON. To control for the effect of caloric surplus, we similarly overfed (+75% energy) the same subjects for three days with a fat-rich diet (78 E% fat; FAT), consisting of primarily unsaturated fatty acids. The three diets were provided in random order. Results After CHO, plasma FGF21 concentration increased 8-fold compared to CON (329 ± 99 vs. 39 ± 9 pg ml−1, p < 0.05). In contrast, after FAT only a non-significant tendency (p = 0.073) to an increase in plasma FGF21 concentration was found. The increase in FGF21 concentration after CHO correlated closely (r = 0.88, p < 0.01) with increased leg glucose uptake (62%, p < 0.05) and increased hepatic glucose production (17%, p < 0.01), indicating increased glucose turnover. Plasma fatty acid (FA) concentration was decreased by 68% (p < 0.01), supported by reduced subcutaneous adipose tissue HSL Ser660 phosphorylation (p < 0.01) and perilipin 1 protein content (p < 0.01), pointing to a suppression of adipose tissue lipolysis. Concomitantly, a 146% increase in the plasma marker of hepatic de novo lipogenesis C16:1 n−7 FA (p < 0.01) was observed together with 101% increased plasma TG concentration (p < 0.001) in association with CHO intake and increased plasma FGF21 concentration. Conclusion Excess dietary carbohydrate, but not fat, led to markedly increased FGF21 secretion in humans, notably without protein restriction, and affected glucose and lipid homeostais. Dietary carbohydrate excess induces circulating FGF21 8-fold in humans. Increased FGF21 was associated with increased hepatic glucose production and lipogenesis. The induction of FGF21 was associated with increased leg glucose uptake. The induction of FGF21 was accompanied by indices of lower adipose tissue lipolysis.
Collapse
Key Words
- AMPK, AMP-activated kinase
- ATGL, adipose triglyceride lipase
- BCA, bicinchoninic acid
- BM, body mass
- BMI, body mass index
- CHO, carbohydrate-rich diet
- CON, control diet
- Carbohydrates
- ChREBP, carbohydrate-responsive element binding protein
- Diet
- FA, fatty acid
- FAT, fat-rich diet
- FGF21
- FGF21, fibroblast growth factor 21
- GLUT4, glucose transporter 4
- HSL, hormone sensitive lipase
- LM, leg mass
- Lipolysis
- Liver
- PKA, protein kinase A
- Ra, rate of appearance
- TG, triacylglycerol
- VLDL, very low density lipoprotein
- VO2peak, maximal oxygen consumption
Collapse
Affiliation(s)
- Anne-Marie Lundsgaard
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Andreas M Fritzen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kim A Sjøberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Lene S Myrmel
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Lise Madsen
- National Institute of Nutrition and Seafood Research, Bergen, Norway; Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
38
|
Sharma S, Taliyan R. Histone deacetylase inhibitors: Future therapeutics for insulin resistance and type 2 diabetes. Pharmacol Res 2016; 113:320-326. [DOI: 10.1016/j.phrs.2016.09.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 12/19/2022]
|
39
|
Hepatic Fgf21 Expression Is Repressed after Simvastatin Treatment in Mice. PLoS One 2016; 11:e0162024. [PMID: 27583452 PMCID: PMC5008788 DOI: 10.1371/journal.pone.0162024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 08/16/2016] [Indexed: 01/16/2023] Open
Abstract
Fibroblast growth factor 21 (Fgf21) is a hormone with emerging beneficial roles in glucose and lipid homeostasis. The interest in Fgf21 as a potential antidiabetic drug and the factors that regulate its production and secretion is growing. Statins are the most widely prescribed drug for the treatment of dyslipidemia. However, the function of statins is not limited to the lowering of cholesterol as they are associated with pleiotropic actions such as antioxidant, anti-inflammatory and cytoprotective effects. The recently described effect of statins on mitochondrial function and the induction of Fgf21 by mitochondrial stress prompted us to investigate the effect of statin treatment on Fgf21 expression in the liver. To this end, C57BL6J male mice and primary mouse hepatocytes were treated with simvastatin, and Fgf21 expression was subsequently assessed by immunoblotting and quantitative real-time PCR. Hepatic Fgf21 protein and mRNA and circulating levels of FGF21significantly decreased in mice that had received simvastatin in their food (0.1% w/w) for 1 week. This effect was also observed with simvastatin doses as low as 0.01% w/w for 1 week or following 2 intraperitoneal injections within a single day. The reduction in Fgf21 mRNA levels was further verified in primary mouse hepatocytes, indicating that the effect of simvastatin is cell autonomous. In conclusion, simvastatin treatment reduced the circulating and hepatic Fgf21 levels and this effect warrants further investigation with reference to its role in metabolism.
Collapse
|
40
|
CREBH-FGF21 axis improves hepatic steatosis by suppressing adipose tissue lipolysis. Sci Rep 2016; 6:27938. [PMID: 27301791 PMCID: PMC4908383 DOI: 10.1038/srep27938] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/26/2016] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue lipolysis produces glycerol and nonesterified fatty acids (NEFA) that serve as energy sources during nutrient scarcity. Adipose tissue lipolysis is tightly regulated and excessive lipolysis causes hepatic steatosis, as NEFA released from adipose tissue constitutes a major source of TG in the liver of patients with nonalcoholic fatty liver diseases. Here we show that the liver-enriched transcription factor CREBH is activated by TG accumulation and induces FGF21, which suppresses adipose tissue lipolysis, ameliorating hepatic steatosis. CREBH-deficient mice developed severe hepatic steatosis due to increased adipose tissue lipolysis, when fasted or fed a high-fat low-carbohydrate ketogenic diet. FGF21 production was impaired in CREBH-deficient mice, and adenoviral overexpression of FGF21 suppressed adipose tissue lipolysis and improved hepatic steatosis in these mice. Thus, our results uncover a negative feedback loop in which CREBH regulates NEFA flux from adipose tissue to the liver via FGF21.
Collapse
|
41
|
Samms RJ, Cheng CC, Kharitonenkov A, Gimeno RE, Adams AC. Overexpression of β-Klotho in Adipose Tissue Sensitizes Male Mice to Endogenous FGF21 and Provides Protection From Diet-Induced Obesity. Endocrinology 2016; 157:1467-80. [PMID: 26901091 DOI: 10.1210/en.2015-1722] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The endocrine hormone fibroblast growth factor 21 (FGF21) is induced in the adaptive response to nutrient deprivation, where it serves to regulate the integrated response to fasting via its primary receptor complex, FGF receptor 1 coupled with the cofactor β-klotho (KLB) in target tissues. Curiously, endogenous FGF21 levels are also elevated in preclinical models of obesity and in obese/diabetic individuals. In addition to higher FGF21 levels, reduced KLB expression in liver and adipose tissue has been noted in these same individuals, suggesting that obesity may represent an FGF21 resistant state. To explore the contribution of tissue-specific KLB levels to endogenous FGF21 activity, in both fasting and high-fat diet feeding conditions, we generated animals overexpressing KLB in liver (LKLBOE) or adipose (ATKLBOE). Supportive of tissue-specific partitioning of FGF21 action, after chronic high-fat feeding, ATKLBOE mice gained significantly less weight than WT. Reduced weight gain was associated with elevated caloric expenditure, accompanied by a reduced respiratory exchange ratio and lower plasma free fatty acids levels, suggestive of augmented lipid metabolism. In contrast, LKLBOE had no effect on body weight but did reduce plasma cholesterol. The metabolic response to fasting was enhanced in LKLBOE mice, evidenced by increased ketone production, whereas no changes in this were noted in ATKLBOE mice. Taken together, these data provide further support that specific effects of FGF21 are mediated via engagement of distinct target organs. Furthermore, enhancing KLB expression in adipose may sensitize to endogenous FGF21, thus representing a novel strategy to combat metabolic disease.
Collapse
Affiliation(s)
- Ricardo J Samms
- Lilly Research Laboratories (R.J.S., C.C.C., R.E.G., A.C.A.) and formerly of Lilly Research Laboratories (A.K.), Lilly Corporate Center, Indianapolis, Indiana 46285
| | - Christine C Cheng
- Lilly Research Laboratories (R.J.S., C.C.C., R.E.G., A.C.A.) and formerly of Lilly Research Laboratories (A.K.), Lilly Corporate Center, Indianapolis, Indiana 46285
| | - Alexei Kharitonenkov
- Lilly Research Laboratories (R.J.S., C.C.C., R.E.G., A.C.A.) and formerly of Lilly Research Laboratories (A.K.), Lilly Corporate Center, Indianapolis, Indiana 46285
| | - Ruth E Gimeno
- Lilly Research Laboratories (R.J.S., C.C.C., R.E.G., A.C.A.) and formerly of Lilly Research Laboratories (A.K.), Lilly Corporate Center, Indianapolis, Indiana 46285
| | - Andrew C Adams
- Lilly Research Laboratories (R.J.S., C.C.C., R.E.G., A.C.A.) and formerly of Lilly Research Laboratories (A.K.), Lilly Corporate Center, Indianapolis, Indiana 46285
| |
Collapse
|
42
|
Donate-Correa J, Martín-Núñez E, Delgado NP, de Fuentes MM, Arduan AO, Mora-Fernández C, Navarro González JF. Implications of Fibroblast growth factor/Klotho system in glucose metabolism and diabetes. Cytokine Growth Factor Rev 2016; 28:71-77. [PMID: 26706229 DOI: 10.1016/j.cytogfr.2015.12.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 12/02/2015] [Accepted: 12/07/2015] [Indexed: 01/03/2023]
Abstract
Diabetes mellitus, especially type 2 diabetes, remains the dominant metabolic disease worldwide, with an expected increase in prevalence of over 50% in the next 20 years. Our knowledge about the pathophysiology of type 2 diabetes continues to be incomplete, with unmet medical need for new therapies. The characterization of the fibroblast growth factor (FGF) family and the discovery of endocrine FGFs provided new information on the mechanisms of regulation and homeostasis of carbohydrate metabolism. More specifically, FGF19 and FGF21 signaling pathways have been linked to different glucose metabolic processes, including hepatic glucose synthesis, glycogen synthesis, glucose uptake, and insulin sensitivity, among others, and these molecules have been further related to the pathophysiology of diabetes mellitus. In-depth comprehension of these growth factors may bring to light new potential therapeutic targets for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Ernesto Martín-Núñez
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Nayra Pérez Delgado
- Clinical Analysis Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Mercedes Muros de Fuentes
- Clinical Analysis Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Alberto Ortiz Arduan
- IIS-Fundacion Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo-IRSIN and REDinREN, Madrid, Spain; Department of Nephrology, Fundación Jiménez Díaz, Madrid, Spain
| | - Carmen Mora-Fernández
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Juan F Navarro González
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; Nephrology Service, University Hospital Nuestra Señora de Candelaria, REDinREN, Santa Cruz de Tenerife, Spain.
| |
Collapse
|
43
|
So WY, Leung PS. Fibroblast Growth Factor 21 As an Emerging Therapeutic Target for Type 2 Diabetes Mellitus. Med Res Rev 2016; 36:672-704. [PMID: 27031294 DOI: 10.1002/med.21390] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/13/2015] [Accepted: 02/15/2016] [Indexed: 12/19/2022]
Abstract
Fibroblast growth factor (FGF) 21 is a distinctive member of the FGF family that functions as an endocrine factor. It is expressed predominantly in the liver, but is also found in adipose tissue and the pancreas. Pharmacological studies have shown that FGF21 normalizes glucose and lipid homeostasis, thereby preventing the development of metabolic disorders, such as obesity and diabetes. Despite growing evidence for the therapeutic potential of FGF21, paradoxical increases of FGF21 in different disease conditions point to the existence of FGF21 resistance. In this review, we give a critical appraisal of recent advances in the understanding of the regulation of FGF21 production under various physiological conditions, its antidiabetic actions, and the clinical implications. We also discuss recent preclinical and clinical trials using engineered FGF21 analogs in the management of diabetes, as well as the potential side effects of FGF21 therapy.
Collapse
Affiliation(s)
- Wing Yan So
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
44
|
Jager J, Wang F, Fang B, Lim HW, Peed LC, Steger DJ, Won KJ, Kharitonenkov A, Adams AC, Lazar MA. The Nuclear Receptor Rev-erbα Regulates Adipose Tissue-specific FGF21 Signaling. J Biol Chem 2016; 291:10867-75. [PMID: 27002153 DOI: 10.1074/jbc.m116.719120] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Indexed: 01/14/2023] Open
Abstract
FGF21 is an atypical member of the FGF family that functions as a hormone to regulate carbohydrate and lipid metabolism. Here we demonstrate that the actions of FGF21 in mouse adipose tissue, but not in liver, are modulated by the nuclear receptor Rev-erbα, a potent transcriptional repressor. Interrogation of genes induced in the absence of Rev-erbα for Rev-erbα-binding sites identified βKlotho, an essential coreceptor for FGF21, as a direct target gene of Rev-erbα in white adipose tissue but not liver. Rev-erbα ablation led to the robust elevated expression of βKlotho. Consequently, the effects of FGF21 were markedly enhanced in the white adipose tissue of mice lacking Rev-erbα. A major Rev-erbα-controlled enhancer at the Klb locus was also bound by the adipocytic transcription factor peroxisome proliferator-activated receptor (PPAR) γ, which regulates its activity in the opposite direction. These findings establish Rev-erbα as a specific modulator of FGF21 signaling in adipose tissue.
Collapse
Affiliation(s)
- Jennifer Jager
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Fenfen Wang
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Bin Fang
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Hee-Woong Lim
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Lindsey C Peed
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David J Steger
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kyoung-Jae Won
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Alexei Kharitonenkov
- the Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, and
| | - Andrew C Adams
- the Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285
| | - Mitchell A Lazar
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104,
| |
Collapse
|
45
|
Abstract
Fibroblast growth factor 21 (FGF21) is a peptide hormone that is synthesized by several organs and regulates energy homeostasis. Excitement surrounding this relatively recently identified hormone is based on the documented metabolic beneficial effects of FGF21, which include weight loss and improved glycemia. The biology of FGF21 is intrinsically complicated owing to its diverse metabolic functions in multiple target organs and its ability to act as an autocrine, paracrine, and endocrine factor. In the liver, FGF21 plays an important role in the regulation of fatty acid oxidation both in the fasted state and in mice consuming a high-fat, low-carbohydrate ketogenic diet. FGF21 also regulates fatty acid metabolism in mice consuming a diet that promotes hepatic lipotoxicity. In white adipose tissue (WAT), FGF21 regulates aspects of glucose metabolism, and in susceptible WAT depots, it can cause browning. This peptide is highly expressed in the pancreas, where it appears to play an anti-inflammatory role in experimental pancreatitis. It also has an anti-inflammatory role in cardiac muscle. Although typically not expressed in skeletal muscle, FGF21 is induced in situations of muscle stress, particularly mitochondrial myopathies. FGF21 has been proposed as a novel therapeutic for metabolic complications such as diabetes and fatty liver disease. This review aims to interpret and delineate the ever-expanding complexity of FGF21 physiology.
Collapse
Affiliation(s)
- Ffolliott Martin Fisher
- Department of Medicine, Harvard Medical School, and Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215;
| | - Eleftheria Maratos-Flier
- Department of Medicine, Harvard Medical School, and Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215;
| |
Collapse
|
46
|
Therapeutic potential of the endocrine fibroblast growth factors FGF19, FGF21 and FGF23. Nat Rev Drug Discov 2015; 15:51-69. [PMID: 26567701 DOI: 10.1038/nrd.2015.9] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The endocrine fibroblast growth factors (FGFs), FGF19, FGF21 and FGF23, are critical for maintaining whole-body homeostasis, with roles in bile acid, glucose and lipid metabolism, modulation of vitamin D and phosphate homeostasis and metabolic adaptation during fasting. Given these functions, the endocrine FGFs have therapeutic potential in a wide array of chronic human diseases, including obesity, type 2 diabetes, cancer, and kidney and cardiovascular disease. However, the safety and feasibility of chronic endocrine FGF administration has been challenged, and FGF analogues and mimetics are now being investigated. Here, we discuss current knowledge of the complex biology of the endocrine FGFs and assess how this may be harnessed therapeutically.
Collapse
|
47
|
Stemmer K, Zani F, Habegger KM, Neff C, Kotzbeck P, Bauer M, Yalamanchilli S, Azad A, Lehti M, Martins PJF, Müller TD, Pfluger PT, Seeley RJ. FGF21 is not required for glucose homeostasis, ketosis or tumour suppression associated with ketogenic diets in mice. Diabetologia 2015; 58:2414-23. [PMID: 26099854 PMCID: PMC5144740 DOI: 10.1007/s00125-015-3668-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 05/26/2015] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Ketogenic diets (KDs) have increasingly gained attention as effective means for weight loss and potential adjunctive treatment of cancer. The metabolic benefits of KDs are regularly ascribed to enhanced hepatic secretion of fibroblast growth factor 21 (FGF21) and its systemic effects on fatty-acid oxidation, energy expenditure (EE) and body weight. Ambiguous data from Fgf21-knockout animal strains and low FGF21 concentrations reported in humans with ketosis have nevertheless cast doubt regarding the endogenous function of FGF21. We here aimed to elucidate the causal role of FGF21 in mediating the therapeutic benefits of KDs on metabolism and cancer. METHODS We established a dietary model of increased vs decreased FGF21 by feeding C57BL/6J mice with KDs, either depleted of protein or enriched with protein. We furthermore used wild-type and Fgf21-knockout mice that were subjected to the respective diets, and monitored energy and glucose homeostasis as well as tumour growth after transplantation of Lewis lung carcinoma cells. RESULTS Hepatic and circulating, but not adipose tissue, FGF21 levels were profoundly increased by protein starvation, independent of the state of ketosis. We demonstrate that endogenous FGF21 is not essential for the maintenance of normoglycaemia upon protein and carbohydrate starvation and is therefore not needed for the effects of KDs on EE. Furthermore, the tumour-suppressing effects of KDs were independent of FGF21 and, rather, driven by concomitant protein and carbohydrate starvation. CONCLUSIONS/INTERPRETATION Our data indicate that the multiple systemic effects of KD exposure in mice, previously ascribed to increased FGF21 secretion, are rather a consequence of protein malnutrition.
Collapse
Affiliation(s)
- Kerstin Stemmer
- Division of Metabolism and Cancer, Institute for Diabetes and Obesity, Helmholtz Centre Munich, Neuherberg, Germany
| | - Fabio Zani
- Division of Metabolism and Cancer, Institute for Diabetes and Obesity, Helmholtz Centre Munich, Neuherberg, Germany
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Department of Medicine-Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christina Neff
- Division of Metabolism and Cancer, Institute for Diabetes and Obesity, Helmholtz Centre Munich, Neuherberg, Germany
| | - Petra Kotzbeck
- Division of Metabolism and Cancer, Institute for Diabetes and Obesity, Helmholtz Centre Munich, Neuherberg, Germany
| | - Michaela Bauer
- Division of Metabolism and Cancer, Institute for Diabetes and Obesity, Helmholtz Centre Munich, Neuherberg, Germany
| | - Suma Yalamanchilli
- Division of Metabolism and Cancer, Institute for Diabetes and Obesity, Helmholtz Centre Munich, Neuherberg, Germany
| | - Ali Azad
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA
| | - Maarit Lehti
- LIKES Research Center for Sport and Health Sciences, Jyväskylä, Finland
| | - Paulo J F Martins
- Division of Hematology-Oncology, Department of Internal Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Timo D Müller
- Division of Molecular Pharmacology, Institute for Diabetes and Obesity, Helmholtz Centre Munich, Neuherberg, Germany
| | - Paul T Pfluger
- Research Unit NeuroBiology of Diabetes, Helmholtz Centre Munich, Neuherberg, Germany
| | - Randy J Seeley
- Department of Surgery, University of Michigan, North Campus Research Center, 2800 Plymouth Road, Ann Arbor, MI, 48109-2800, USA.
| |
Collapse
|
48
|
Abstract
Many organisms have developed a robust ability to adapt and survive in the face of environmental perturbations that threaten the integrity of their genome, proteome, or metabolome. Studies in multiple model organisms have shown that, in general, when exposed to stress, cells activate a complex prosurvival signaling network that includes immune and DNA damage response genes, chaperones, antioxidant enzymes, structural proteins, metabolic enzymes, and noncoding RNAs. The manner of activation runs the gamut from transcriptional induction of genes to increased stability of transcripts to posttranslational modification of important biosynthetic proteins within the stressed tissue. Superimposed on these largely autonomous effects are nonautonomous responses in which the stressed tissue secretes peptides and other factors that stimulate tissues in different organs to embark on processes that ultimately help the organism as a whole cope with stress. This review focuses on the mechanisms by which tissues in one organ adapt to environmental challenges by regulating stress responses in tissues of different organs.
Collapse
Affiliation(s)
- Edward Owusu-Ansah
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032;
| | | |
Collapse
|
49
|
Tanaka N, Takahashi S, Zhang Y, Krausz KW, Smith PB, Patterson AD, Gonzalez FJ. Role of fibroblast growth factor 21 in the early stage of NASH induced by methionine- and choline-deficient diet. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1852:1242-1252. [PMID: 25736301 PMCID: PMC4433820 DOI: 10.1016/j.bbadis.2015.02.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/18/2015] [Accepted: 02/24/2015] [Indexed: 02/06/2023]
Abstract
Fibroblast growth factor 21 (FGF21) is a modulator of energy homeostasis and is increased in human nonalcoholic liver disease (NAFLD) and after feeding of methionine- and choline-deficient diet (MCD), a conventional inducer of murine nonalcoholic steatohepatitis (NASH). However, the significance of FGF21 induction in the occurrence of MCD-induced NASH remains undetermined. C57BL/6J Fgf21-null and wild-type mice were treated with MCD for 1 week. Hepatic Fgf21 mRNA was increased early after commencing MCD treatment independent of peroxisome proliferator-activated receptor (PPAR) α and farnesoid X receptor. While no significant differences in white adipose lipolysis were seen in both genotypes, hepatic triglyceride (TG) contents were increased in Fgf21-null mice, likely due to the up-regulation of genes encoding CD36 and phosphatidic acid phosphatase 2a/2c, involved in fatty acid (FA) uptake and diacylglycerol synthesis, respectively, and suppression of increased mRNAs encoding carnitine palmitoyl-CoA transferase 1α, PPARγ coactivator 1α, and adipose TG lipase, which are associated with lipid clearance in the liver. The MCD-treated Fgf21-null mice showed increased hepatic endoplasmic reticulum (ER) stress. Exposure of primary hepatocytes to palmitic acid elevated the mRNA levels encoding DNA damage-inducible transcript 3, an indicator of ER stress, and FGF21 in a PPARα-independent manner, suggesting that lipid-induced ER stress can enhance hepatic FGF21 expression. Collectively, FGF21 is elevated in the early stage of MCD-induced NASH likely to minimize hepatic lipid accumulation and ensuing ER stress. These results provide a possible mechanism on how FGF21 is increased in NAFLD/NASH.
Collapse
Affiliation(s)
- Naoki Tanaka
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Shogo Takahashi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yuan Zhang
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Philip B Smith
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, United States
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
50
|
Douris N, Stevanovic DM, Fisher FM, Cisu TI, Chee MJ, Nguyen NL, Zarebidaki E, Adams AC, Kharitonenkov A, Flier JS, Bartness TJ, Maratos-Flier E. Central Fibroblast Growth Factor 21 Browns White Fat via Sympathetic Action in Male Mice. Endocrinology 2015; 156:2470-81. [PMID: 25924103 PMCID: PMC4475718 DOI: 10.1210/en.2014-2001] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fibroblast growth factor 21 (FGF21) has multiple metabolic actions, including the induction of browning in white adipose tissue. Although FGF21 stimulated browning results from a direct interaction between FGF21 and the adipocyte, browning is typically associated with activation of the sympathetic nervous system through cold exposure. We tested the hypothesis that FGF21 can act via the brain, to increase sympathetic activity and induce browning, independent of cell-autonomous actions. We administered FGF21 into the central nervous system via lateral ventricle infusion into male mice and found that the central treatment increased norepinephrine turnover in target tissues that include the inguinal white adipose tissue and brown adipose tissue. Central FGF21 stimulated browning as assessed by histology, expression of uncoupling protein 1, and the induction of gene expression associated with browning. These effects were markedly attenuated when mice were treated with a β-blocker. Additionally, neither centrally nor peripherally administered FGF21 initiated browning in mice lacking β-adrenoceptors, demonstrating that an intact adrenergic system is necessary for FGF21 action. These data indicate that FGF21 can signal in the brain to activate the sympathetic nervous system and induce adipose tissue thermogenesis.
Collapse
MESH Headings
- Adipocytes, Brown/metabolism
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adrenergic beta-Antagonists/pharmacology
- Animals
- Fibroblast Growth Factors/pharmacology
- Infusions, Intraventricular
- Ion Channels/drug effects
- Ion Channels/metabolism
- Male
- Mice
- Mice, Knockout
- Mitochondrial Proteins/drug effects
- Mitochondrial Proteins/metabolism
- Receptors, Adrenergic, beta/genetics
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-3/genetics
- Sympathetic Nervous System/drug effects
- Sympathetic Nervous System/metabolism
- Thermogenesis
- Uncoupling Protein 1
Collapse
Affiliation(s)
- Nicholas Douris
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Darko M Stevanovic
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Ffolliott M Fisher
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Theodore I Cisu
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Melissa J Chee
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Ngoc L Nguyen
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Eleen Zarebidaki
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Andrew C Adams
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Alexei Kharitonenkov
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Jeffrey S Flier
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Timothy J Bartness
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Eleftheria Maratos-Flier
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| |
Collapse
|