5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      FGF21 protects against hepatic lipotoxicity and macrophage activation to attenuate fibrogenesis in nonalcoholic steatohepatitis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Analogues of the hepatokine fibroblast growth factor 21 (FGF21) are in clinical development for type 2 diabetes and nonalcoholic steatohepatitis (NASH) treatment. Although their glucose-lowering and insulin-sensitizing effects have been largely unraveled, the mechanisms by which they alleviate liver injury have only been scarcely addressed. Here, we aimed to unveil the mechanisms underlying the protective effects of FGF21 on NASH using APOE*3-Leiden.CETP mice, a well-established model for human-like metabolic diseases. Liver-specific FGF21 overexpression was achieved in mice, followed by administration of a high-fat high-cholesterol diet for 23 weeks. FGF21 prevented hepatic lipotoxicity, accompanied by activation of thermogenic tissues and attenuation of adipose tissue inflammation, improvement of hyperglycemia and hypertriglyceridemia, and upregulation of hepatic programs involved in fatty acid oxidation and cholesterol removal. Furthermore, FGF21 inhibited hepatic inflammation, as evidenced by reduced Kupffer cell (KC) activation, diminished monocyte infiltration, and lowered accumulation of monocyte-derived macrophages. Moreover, FGF21 decreased lipid- and scar-associated macrophages, which correlated with less hepatic fibrosis as demonstrated by reduced collagen accumulation. Collectively, hepatic FGF21 overexpression limits hepatic lipotoxicity, inflammation, and fibrogenesis. Mechanistically, FGF21 blocks hepatic lipid influx and accumulation through combined endocrine and autocrine signaling, respectively, which prevents KC activation and lowers the presence of lipid- and scar-associated macrophages to inhibit fibrogenesis.

          eLife digest

          High-calorie modern diets have contributed to growing rates of obesity-linked diseases. One such disease is non-alcoholic steatohepatitis or NASH for short, which affects about 5% of adults in the United States. The livers of people with this condition accumulate fat, become inflamed, and develop scar tissue. People with NASH are also at increased risk of developing liver cancer, type 2 diabetes, and heart disease. Currently, no drugs are available to treat the condition and prevent such severe complications.

          Previous research has shown the liver produces a stress hormone, called FGF21, in response to fat accumulation. This hormone boosts fat burning and so helps to reduce excess fat in the liver. Drugs that mimic FGF21 have already been developed for type 2 diabetes. But so far, it was unclear if such drugs could also help reduce liver inflammation and scarring in patients with NASH.

          Liu et al. show that increasing the production of FGF21 in mice with a NASH-like condition reduces fat accumulation, liver inflammation, and scarring. In the experiments, the researchers used gene therapy to ramp up FGF21 production in the livers of mice that develop obesity and a NASH-like condition when fed a high-fat diet for 23 weeks. Increasing FGF21 production prevented the mice from developing obesity while on the high fat diet by making the body burn more fat in the liver and brown fat tissue. The treatment also reduced inflammation and prevented scarring by reducing the number and activity of immune cells in the liver.

          Increasing the production of the stress hormone FGF21 prevents diet-induced obesity and NASH in mice fed a high-fat diet. More studies are necessary to determine if using gene therapy to increase FGF21 may also cause weight loss and could reverse liver damage in mice that already have NASH. If this approach is effective in mice, it may be tested in humans, a process that may take several years. If human studies are successful, FGF21-boosting therapy might provide a new treatment approach for obesity or NASH.

          Related collections

          Most cited references67

          • Record: found
          • Abstract: not found
          • Article: not found

          A RAPID METHOD OF TOTAL LIPID EXTRACTION AND PURIFICATION

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mechanisms of NAFLD development and therapeutic strategies

            There has been a rise in the prevalence of nonalcoholic fatty liver disease (NAFLD), paralleling a worldwide increase in diabetes and metabolic syndrome. NAFLD, a continuum of liver abnormalities from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH), has a variable course but can lead to cirrhosis and liver cancer. Here we review the pathogenic and clinical features of NAFLD, its major comorbidities, clinical progression and risk of complications and in vitro and animal models of NAFLD enabling refinement of therapeutic targets that can accelerate drug development. We also discuss evolving principles of clinical trial design to evaluate drug efficacy and the emerging targets for drug development that involve either single agents or combination therapies intended to arrest or reverse disease progression.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Resolving the fibrotic niche of human liver cirrhosis at single cell level

              Currently there are no effective antifibrotic therapies for liver cirrhosis, a major killer worldwide. To obtain a cellular resolution of directly-relevant pathogenesis and to inform therapeutic design, we profile the transcriptomes of over 100,000 human single cells, yielding molecular definitions for non-parenchymal cell types present in healthy and cirrhotic human liver. We uncover a novel scar-associated TREM2+CD9+ macrophage subpopulation, which expands in liver fibrosis, differentiates from circulating monocytes and is pro-fibrogenic. We also define novel ACKR1+ and PLVAP+ endothelial cells which expand in cirrhosis, are topographically scar-restricted and enhance leucocyte transmigration. Multi-lineage ligand-receptor modelling of interactions between the novel scar-associated macrophages, endothelial cells and PDGFRα+ collagen-producing mesenchymal cells reveals intra-scar activity of several pro-fibrogenic pathways including TNFRSF12A, PDGFR and NOTCH signalling. Our work dissects unanticipated aspects of the cellular and molecular basis of human organ fibrosis at a single-cell level, and provides the conceptual framework required to discover rational therapeutic targets in liver cirrhosis.
                Bookmark

                Author and article information

                Contributors
                Role: Reviewing Editor
                Role: Senior Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                17 January 2023
                2023
                : 12
                : e83075
                Affiliations
                [1 ] Department of Medicine, Division of Endocrinology, Leiden University Medical Center ( https://ror.org/05xvt9f17) Leiden Netherlands
                [2 ] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center ( https://ror.org/05xvt9f17) Leiden Netherlands
                [3 ] Department of Parasitology, Leiden University Medical Center ( https://ror.org/05xvt9f17) Leiden Netherlands
                [4 ] Department of Cell and Chemical Biology, Leiden University Medical Center ( https://ror.org/05xvt9f17) Leiden Netherlands
                [5 ] Department of Gastroenterology and Hepatology, Leiden University Medical Center ( https://ror.org/05xvt9f17) Leiden Netherlands
                [6 ] Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca ( https://ror.org/04wwrrg31) Gothenburg Sweden
                [7 ] Biologics Engineering and Targeted Delivery, Oncology R&D, AstraZeneca ( https://ror.org/043cec594) Gaithersburg United States
                [8 ] Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca ( https://ror.org/043cec594) Gaithersburg United States
                [9 ] Med-X institute, Center for Immunological and Metabolic Diseases, and Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University ( https://ror.org/017zhmm22) Xi'an China
                Yale School of Medicine ( https://ror.org/03v76x132) United States
                Icahn School of Medicine at Mount Sinai ( https://ror.org/04a9tmd77) United States
                Yale School of Medicine ( https://ror.org/03v76x132) United States
                Yale School of Medicine ( https://ror.org/03v76x132) United States
                Author information
                https://orcid.org/0000-0002-2852-8953
                https://orcid.org/0000-0002-3739-4934
                https://orcid.org/0000-0002-8323-0658
                https://orcid.org/0000-0002-3231-2733
                https://orcid.org/0000-0002-8455-4988
                Article
                83075
                10.7554/eLife.83075
                9928421
                36648330
                25555187-f4ed-4dcf-90a6-ffd5c29278cf
                © 2023, Liu et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 30 August 2022
                : 16 January 2023
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100003092, Diabetes Fonds;
                Award ID: 2015.81.1808
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100003246, Netherlands Organisation for Scientific Research;
                Award ID: VENI grant 91617027
                Award Recipient :
                Funded by: Chinese Scholarship Council;
                Award ID: CSC 201606010321
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100009708, Novo Nordisk Foundation;
                Award ID: NNF18OC0032394
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100002996, Hartstichting;
                Award ID: The Netherlands Cardiovascular Research Initiative CVON-GENIUS-2
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Immunology and Inflammation
                Medicine
                Custom metadata
                Fibroblast growth factor 21 blocks hepatic lipid influx and accumulation, prevents Kupffer cell activation, and inhibits the formation of hepatic lipid- and scar-associated macrophages, thereby likely inhibiting fibrogenesis in nonalcoholic steatohepatitis in humanized APOE*3-Leiden.CETP mice.

                Life sciences
                fibroblast growth factor 21,steatohepatitis,lipid/scar-associated macrophages,liver-adipose tissue crosstalk,mouse

                Comments

                Comment on this article