10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Modulating sphingosine 1-phosphate receptor signaling skews intrahepatic leukocytes and attenuates murine 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

          Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid associated with nonalcoholic steatohepatitis (NASH). Immune cell-driven inflammation is a key determinant of NASH progression. Macrophages, monocytes, NK cells, T cells, NKT cells, and B cells variably express S1P receptors from a repertoire of 5 receptors termed S1P 1 – S1P 5. We have previously demonstrated that non-specific S1P receptor antagonism ameliorates NASH and attenuates hepatic macrophage accumulation. However, the effect of S1P receptor antagonism on additional immune cell populations in NASH remains unknown. We hypothesized that S1P receptor specific modulation may ameliorate NASH by altering leukocyte recruitment. A murine NASH model was established by dietary feeding of C57BL/6 male mice with a diet high in fructose, saturated fat, and cholesterol (FFC) for 24 weeks. In the last 4 weeks of dietary feeding, the mice received the S1P 1,4,5 modulator Etrasimod or the S1P 1 modulator Amiselimod, daily by oral gavage. Liver injury and inflammation were determined by histological and gene expression analyses. Intrahepatic leukocyte populations were analyzed by flow cytometry, immunohistochemistry, and mRNA expression. Alanine aminotransferase, a sensitive circulating marker for liver injury, was reduced in response to Etrasimod and Amiselimod treatment. Liver histology showed a reduction in inflammatory foci in Etrasimod-treated mice. Etrasimod treatment substantially altered the intrahepatic leukocyte populations through a reduction in the frequency of T cells, B cells, and NKT cells and a proportional increase in CD11b + myeloid cells, polymorphonuclear cells, and double negative T cells in FFC-fed and control standard chow diet (CD)-fed mice. In contrast, FFC-fed Amiselimod-treated mice showed no changes in the frequencies of intrahepatic leukocytes. Consistent with the improvement in liver injury and inflammation, hepatic macrophage accumulation and the gene expression of proinflammatory markers such as Lgals3 and Mcp-1 were decreased in Etrasimod-treated FFC-fed mice. Etrasimod treated mouse livers demonstrated an increase in non-inflammatory ( Marco) and lipid associated ( Trem2) macrophage markers. Thus, S1P 1,4,5 modulation by Etrasimod is more effective than S1P 1 antagonism by Amiselimod, at the dose tested, in ameliorating NASH, likely due to the alteration of leukocyte trafficking and recruitment. Etrasimod treatment results in a substantial attenuation of liver injury and inflammation in murine NASH.

          Related collections

          Most cited references38

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

          Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris

          (2018)
          We have created a compendium of single cell transcriptomic data from the model organism Mus musculus comprising more than 100,000 cells from 20 organs and tissues. These data represent a new resource for cell biology, reveal gene expression in poorly characterized cell populations, and allow for direct and controlled comparison of gene expression in cell types shared between tissues, such as T-lymphocytes and endothelial cells from different anatomical locations. Two distinct technical approaches were used for most organs: one approach, microfluidic droplet-based 3’-end counting, enabled the survey of thousands of cells at relatively low coverage, while the other, FACS-based full length transcript analysis, enabled characterization of cell types with high sensitivity and coverage. The cumulative data provide the foundation for an atlas of transcriptomic cell biology.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner

            Immune cells residing in white adipose tissue have been highlighted as important factors contributing to the pathogenesis of metabolic diseases, but the molecular regulators that drive adipose tissue immune cell remodeling during obesity remain largely unknown. Using index and transcriptional single-cell sorting, we comprehensively map all adipose tissue immune populations in both mice and humans during obesity. We describe a novel and conserved Trem2 + lipid-associated macrophage (LAM) subset and identify markers, spatial localization, origin, and functional pathways associated with these cells. Genetic ablation of Trem2 in mice globally inhibits the downstream molecular LAM program, leading to adipocyte hypertrophy as well as systemic hypercholesterolemia, body fat accumulation, and glucose intolerance. These findings identify Trem2 signaling as a major pathway by which macrophages respond to loss of tissue-level lipid homeostasis, highlighting Trem2 as a key sensor of metabolic pathologies across multiple tissues and a potential therapeutic target in metabolic diseases.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Emerging biology of sphingosine-1-phosphate: its role in pathogenesis and therapy.

              Membrane sphingolipids are metabolized to sphingosine-1-phosphate (S1P), a bioactive lipid mediator that regulates many processes in vertebrate development, physiology, and pathology. Once exported out of cells by cell-specific transporters, chaperone-bound S1P is spatially compartmentalized in the circulatory system. Extracellular S1P interacts with five GPCRs that are widely expressed and transduce intracellular signals to regulate cellular behavior, such as migration, adhesion, survival, and proliferation. While many organ systems are affected, S1P signaling is essential for vascular development, neurogenesis, and lymphocyte trafficking. Recently, a pharmacological S1P receptor antagonist has won approval to control autoimmune neuroinflammation in multiple sclerosis. The availability of pharmacological tools as well as mouse genetic models has revealed several physiological actions of S1P and begun to shed light on its pathological roles. The unique mode of signaling of this lysophospholipid mediator is providing novel opportunities for therapeutic intervention, with possibilities to target not only GPCRs but also transporters, metabolic enzymes, and chaperones.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                21 April 2023
                2023
                : 14
                : 1130184
                Affiliations
                [1] 1 Division of Gastroenterology and Hepatology, Mayo Clinic , Rochester, MN, United States
                [2] 2 Department of Integrative Biology and Physiology, University of Minnesota , Minneapolis, MN, United States
                [3] 3 Department of Internal Medicine, College of Medicine, The Catholic University of Korea , Seoul, Republic of Korea
                [4] 4 Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester, MN, United States
                Author notes

                Edited by: Yongzhan Nie, Fourth Military Medical University, China

                Reviewed by: Peter Darlington, Concordia University, Canada; Rebecca L. McCullough, University of Colorado Anschutz Medical Campus, United States

                *Correspondence: Harmeet Malhi, malhi.harmeet@ 123456mayo.edu

                †Present address: Debanjali Dasgupta, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States

                This article was submitted to Inflammation, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2023.1130184
                10160388
                37153573
                fdb0c8f5-5af3-4ecc-ac97-fdc60a36a756
                Copyright © 2023 Liao, Barrow, Venkatesan, Nakao, Mauer, Fredrickson, Song, Sehrawat, Dasgupta, Graham, Revelo and Malhi

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 23 December 2022
                : 10 April 2023
                Page count
                Figures: 6, Tables: 2, Equations: 0, References: 38, Pages: 14, Words: 6029
                Funding
                Funded by: National Institutes of Health , doi 10.13039/100000002;
                Award ID: DK-111378, DK122056
                Funded by: Mayo Foundation for Medical Education and Research , doi 10.13039/100007048;
                This work is supported by DK111378 (HM) and DK122056 (XR), the Mayo Clinic Center for Cell Signaling (P30DK084567), the Mayo Foundation (HM) and the Microscopy Core and Histology Core of Mayo Clinic.
                Categories
                Immunology
                Original Research

                Immunology
                lipotoxicity,fatty liver,sphingolipids,etrasimod,amiselimod
                Immunology
                lipotoxicity, fatty liver, sphingolipids, etrasimod, amiselimod

                Comments

                Comment on this article