24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Sphingosine-1-Phosphate Prevents Egress of Hematopoietic Stem Cells From Liver to Reduce Fibrosis

      research-article
      1 , , 1 , 2 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 1 ,   3 , 3 , 2 , 4 , 1 , 1 ,
      Gastroenterology
      W.B. Saunders
      Mouse Model, CCR2, Sphingolipid, Immune Cell Localization, α-SMA, α-smooth muscle actin, APC, allophycocyanin, BM, bone marrow, CCL, chemokine (C-C motif) ligand, CFSE, carboxyfluorescein succinimidyl ester, CLP, common lymphoid progenitors, CMP, common myeloid progenitors, DiR, 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindotricarbocyanine iodide, HPC-7, hematopoietic progenitor cell line, HSC, hematopoietic stem cell, KSL, c-kit+ sca-1+ and lineageneg, MMP, matrix metalloproteinase, PSR, picrosirius red, SGPL1, sphingosine-1-phosphate lyase, SGPP1, sphingosine-1-phosphate phosphatase, S1P, sphingosine 1-phosphate, SphK1, sphingosine kinase 1

      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

          Background & Aims

          There is growing interest in the use of bone marrow cells to treat liver fibrosis, however, little is known about their antifibrotic efficacy or the identity of their effector cell(s). Sphingosine-1-phosphate (S1P) mediates egress of immune cells from the lymphoid organs into the lymphatic vessels; we investigated its role in the response of hematopoietic stem cells (HSCs) to liver fibrosis in mice.

          Methods

          Purified (c-kit+/sca1+/lin-) HSCs were infused repeatedly into mice undergoing fibrotic liver injury. Chronic liver injury was induced in BoyJ mice by injection of carbon tetrachloride (CCl 4) or placement on a methionine-choline–deficient diet. Some mice were irradiated and given transplants of bone marrow cells from C57BL6 mice, with or without the S1P antagonist FTY720; we then studied HSC mobilization and localization. Migration of HSC lines was quantified in Transwell assays. Levels of S1P in liver, bone marrow, and lymph fluid were measured using an enzyme-linked immunosorbent assay. Liver tissues were collected and analyzed by immunohistochemical quantitative polymerase chain reaction and sphingosine kinase activity assays. We performed quantitative polymerase chain reaction analyses of the expression of sphingosine kinase 1 and 2, sphingosine-1-phosphate lyase 1, and sphingosine-1-phosphate phosphatase 1 in normal human liver and cirrhotic liver from patients with alcohol-related liver disease (n = 6).

          Results

          Infusions of HSCs into mice with liver injury reduced liver scarring based on picrosirius red staining (49.7% reduction in mice given HSCs vs control mice; P < .001), and hepatic hydroxyproline content (328 mg/g in mice given HSCs vs 428 mg/g in control mice; P < .01). HSC infusion also reduced hepatic expression of α-smooth muscle actin (0.19 ± 0.007-fold compared with controls; P < .0001) and collagen type I α 1 chain (0.29 ± 0.17-fold compared with controls; P < .0001). These antifibrotic effects were maintained with infusion of lymphoid progenitors that lack myeloid potential and were associated with increased numbers of recipient neutrophils and macrophages in liver. In studies of HSC cell lines, we found HSCs to recruit monocytes, and this process to require C-C motif chemokine receptor 2. In fibrotic liver tissue from mice and patients, hepatic S1P levels increased owing to increased hepatic sphingosine kinase-1 expression, which contributed to a reduced liver:lymph S1P gradient and limited HSC egress from the liver. Mice given the S1P antagonist (FTY720) with HSCs had increased hepatic retention of HSCs (1697 ± 247 cells in mice given FTY720 vs 982 ± 110 cells in controls; P < .05), and further reductions in fibrosis.

          Conclusions

          In studies of mice with chronic liver injury, we showed the antifibrotic effects of repeated infusions of purified HSCs. We found that HSCs promote recruitment of endogenous macrophages and neutrophils. Strategies to reduce SIP signaling and increase retention of HSCs in the liver could increase their antifibrotic activities and be developed for treatment of patients with liver fibrosis.

          Related collections

          Most cited references22

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

          Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate.

          Lymphocytes require sphingosine-1-phosphate (S1P) receptor-1 to exit lymphoid organs, but the source(s) of extracellular S1P and whether S1P directly promotes egress are unknown. By using mice in which the two kinases that generate S1P were conditionally ablated, we find that plasma S1P is mainly hematopoietic in origin, with erythrocytes a major contributor, whereas lymph S1P is from a distinct radiation-resistant source. Lymphocyte egress from thymus and secondary lymphoid organs was markedly reduced in kinase-deficient mice. Restoration of S1P to plasma rescued egress to blood but not lymph, and the rescue required lymphocyte expression of S1P-receptor-1. Thus, separate sources provide S1P to plasma and lymph to help lymphocytes exit the low-S1P environment of lymphoid organs. Disruption of compartmentalized S1P signaling is a plausible mechanism by which S1P-receptor-1 agonists function as immunosuppressives.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair.

            Macrophages perform both injury-inducing and repair-promoting tasks in different models of inflammation, leading to a model of macrophage function in which distinct patterns of activation have been proposed. We investigated macrophage function mechanistically in a reversible model of liver injury in which the injury and recovery phases are distinct. Carbon tetrachloride---induced liver fibrosis revealed scar-associated macrophages that persisted throughout recovery. A transgenic mouse (CD11b-DTR) was generated in which macrophages could be selectively depleted. Macrophage depletion when liver fibrosis was advanced resulted in reduced scarring and fewer myofibroblasts. Macrophage depletion during recovery, by contrast, led to a failure of matrix degradation. These data provide the first clear evidence that functionally distinct subpopulations of macrophages exist in the same tissue and that these macrophages play critical roles in both the injury and recovery phases of inflammatory scarring.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Bone marrow as a potential source of hepatic oval cells.

              Bone marrow stem cells develop into hematopoietic and mesenchymal lineages but have not been known to participate in production of hepatocytes, biliary cells, or oval cells during liver regeneration. Cross-sex or cross-strain bone marrow and whole liver transplantation were used to trace the origin of the repopulating liver cells. Transplanted rats were treated with 2-acetylaminofluorene, to block hepatocyte proliferation, and then hepatic injury, to induce oval cell proliferation. Markers for Y chromosome, dipeptidyl peptidase IV enzyme, and L21-6 antigen were used to identify liver cells of bone marrow origin. From these cells, a proportion of the regenerated hepatic cells were shown to be donor-derived. Thus, a stem cell associated with the bone marrow has epithelial cell lineage capability.
                Bookmark

                Author and article information

                Contributors
                Journal
                Gastroenterology
                Gastroenterology
                Gastroenterology
                W.B. Saunders
                0016-5085
                1528-0012
                1 July 2017
                July 2017
                : 153
                : 1
                : 233-248.e16
                Affiliations
                [1 ]Birmingham Liver Biomedical Research Unit, National Institute for Health Research, Centre for Liver Research, University of Birmingham, Birmingham, United Kingdom
                [2 ]Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
                [4 ]School Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
                [3 ]Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
                Author notes
                [] Reprint requests Address requests for reprints to: Philip Newsome, MD, PhD, or Andrew King, MD, PhD, Birmingham Liver Biomedical Research Unit, National Institute for Health Research, Centre for Liver Research, Institute of Biomedical Research, 5th Floor, University of Birmingham, Birmingham, B15 2TT United Kingdom. fax: (44) 121-415-8701.Birmingham Liver Biomedical Research UnitNational Institute for Health ResearchCentre for Liver ResearchInstitute of Biomedical Research5th Floor, University of BirminghamBirminghamB15 2TT United Kingdom andyking@ 123456doctors.org.uk P.N.Newsome@ 123456bham.ac.uk
                Article
                S0016-5085(17)30331-1
                10.1053/j.gastro.2017.03.022
                5511862
                28363640
                99094346-7a79-4f6e-b14f-0b03f5bf36f7
                © 2017 The AGA Institute All rights reserved.

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 12 July 2016
                : 18 March 2017
                Categories
                Original Research
                Full Report: Basic and Translational—Liver

                Gastroenterology & Hepatology
                mouse model,ccr2,sphingolipid,immune cell localization,α-sma, α-smooth muscle actin,apc, allophycocyanin,bm, bone marrow,ccl, chemokine (c-c motif) ligand,cfse, carboxyfluorescein succinimidyl ester,clp, common lymphoid progenitors,cmp, common myeloid progenitors,dir, 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindotricarbocyanine iodide,hpc-7, hematopoietic progenitor cell line,hsc, hematopoietic stem cell,ksl, c-kit+ sca-1+ and lineageneg,mmp, matrix metalloproteinase,psr, picrosirius red,sgpl1, sphingosine-1-phosphate lyase,sgpp1, sphingosine-1-phosphate phosphatase,s1p, sphingosine 1-phosphate,sphk1, sphingosine kinase 1

                Comments

                Comment on this article