4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Metabolipidomic profiling reveals an age‐related deficiency of skeletal muscle pro‐resolving mediators that contributes to maladaptive tissue remodeling

      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

          Specialized pro‐resolving mediators actively limit inflammation and support tissue regeneration, but their role in age‐related muscle dysfunction has not been explored. We profiled the mediator lipidome of aging muscle via liquid chromatography‐tandem mass spectrometry and tested whether treatment with the pro‐resolving mediator resolvin D1 (RvD1) could rejuvenate the regenerative ability of aged muscle. Aged mice displayed chronic muscle inflammation and this was associated with a basal deficiency of pro‐resolving mediators 8‐oxo‐RvD1, resolvin E3, and maresin 1, as well as many anti‐inflammatory cytochrome P450‐derived lipid epoxides. Following muscle injury, young and aged mice produced similar amounts of most pro‐inflammatory eicosanoid metabolites of cyclooxygenase (e.g., prostaglandin E 2) and 12‐lipoxygenase (e.g., 12‐hydroxy‐eicosatetraenoic acid), but aged mice produced fewer markers of pro‐resolving mediators including the lipoxins (15‐hydroxy‐eicosatetraenoic acid), D‐resolvins/protectins (17‐hydroxy‐docosahexaenoic acid), E‐resolvins (18‐hydroxy‐eicosapentaenoic acid), and maresins (14‐hydroxy‐docosahexaenoic acid). Similar absences of downstream pro‐resolving mediators including lipoxin A 4, resolvin D6, protectin D1/DX, and maresin 1 in aged muscle were associated with greater inflammation, impaired myofiber regeneration, and delayed recovery of strength. Daily intraperitoneal injection of RvD1 had minimal impact on intramuscular leukocyte infiltration and myofiber regeneration but suppressed inflammatory cytokine expression, limited fibrosis, and improved recovery of muscle function. We conclude that aging results in deficient local biosynthesis of specialized pro‐resolving mediators in muscle and that immunoresolvents may be attractive novel therapeutics for the treatment of muscular injuries and associated pain in the elderly, due to positive effects on recovery of muscle function without the negative side effects on tissue regeneration of non‐steroidal anti‐inflammatory drugs.

          Abstract

          Chronic low‐grade inflammation of aging muscle was associated with a basal deficiency of maresin 1, resolvin E3, 8‐oxo‐resolvin D1, and anti‐inflammatory fatty acid epoxides. Aged mice produced normal amounts of most prostaglandins following muscle injury but were deficient in local biosynthesis of markers of the lipoxins, E‐resolvins, D‐resolvins, and maresins. Systemic treatment with resolvin D1 suppressed inflammatory cytokine expression, limited muscle fibrosis, and improved functional recovery.

          Related collections

          Most cited references45

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

          Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis

          Macrophages (MPs) are important for skeletal muscle regeneration in vivo and may exert beneficial effects on myogenic cell growth through mitogenic and antiapoptotic activities in vitro. However, MPs are highly versatile and may exert various, and even opposite, functions depending on their activation state. We studied monocyte (MO)/MP phenotypes and functions during skeletal muscle repair. Selective labeling of circulating MOs by latex beads in CX3CR1GFP/+ mice showed that injured muscle recruited only CX3CR1lo/Ly-6C+ MOs from blood that exhibited a nondividing, F4/80lo, proinflammatory profile. Then, within muscle, these cells switched their phenotype to become proliferating antiinflammatory CX3CR1hi/Ly-6C− cells that further differentiated into F4/80hi MPs. In vitro, phagocytosis of muscle cell debris induced a switch of proinflammatory MPs toward an antiinflammatory phenotype releasing transforming growth factor β1. In co-cultures, inflammatory MPs stimulated myogenic cell proliferation, whereas antiinflammatory MPs exhibited differentiating activity, assessed by both myogenin expression and fusion into myotubes. Finally, depletion of circulating MOs in CD11b–diphtheria toxin receptor mice at the time of injury totally prevented muscle regeneration, whereas depletion of intramuscular F4/80hi MPs at later stages reduced the diameter of regenerating fibers. In conclusion, injured skeletal muscle recruits MOs exhibiting inflammatory profiles that operate phagocytosis and rapidly convert to antiinflammatory MPs that stimulate myogenesis and fiber growth.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Maresins: novel macrophage mediators with potent antiinflammatory and proresolving actions

            The endogenous cellular and molecular mechanisms that control acute inflammation and its resolution are of wide interest. Using self-resolving inflammatory exudates and lipidomics, we have identified a new pathway involving biosynthesis of potent antiinflammatory and proresolving mediators from the essential fatty acid docosahexaenoic acid (DHA) by macrophages (MΦs). During the resolution of mouse peritonitis, exudates accumulated both 17-hydroxydocosahexaenoic acid, a known marker of 17S-D series resolvin (Rv) and protectin biosynthesis, and 14S-hydroxydocosa-4Z,7Z,10Z,12E,16Z,19Z-hexaenoic acid from endogenous DHA. Addition of either DHA or 14S-hydroperoxydocosa-4Z,7Z,10Z,12E,16Z,19Z-hexaenoic acid to activated MΦs converted these substrates to novel dihydroxy-containing products that possessed potent antiinflammatory and proresolving activity with a potency similar to resolvin E1, 5S,12R,18R-trihydroxyeicosa-6Z,8E,10E,14Z,16E-pentaenoic acid, and protectin D1, 10R,17S-dihydroxydocosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid. Stable isotope incorporation, intermediate trapping, and characterization of physical and biological properties of the products demonstrated a novel 14-lipoxygenase pathway, generating bioactive 7,14-dihydroxydocosa-4Z,8,10,12,16Z,19Z-hexaenoic acid, coined MΦ mediator in resolving inflammation (maresin), which enhances resolution. These findings suggest that maresins and this new metabolome may be involved in some of the beneficial actions of DHA and MΦs in tissue homeostasis, inflammation resolution, wound healing, and host defense.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The central role of muscle stem cells in regenerative failure with aging.

              Skeletal muscle mass, function, and repair capacity all progressively decline with aging, restricting mobility, voluntary function, and quality of life. Skeletal muscle repair is facilitated by a population of dedicated muscle stem cells (MuSCs), also known as satellite cells, that reside in anatomically defined niches within muscle tissues. In adult tissues, MuSCs are retained in a quiescent state until they are primed to regenerate damaged muscle through cycles of self-renewal divisions. With aging, muscle tissue homeostasis is progressively disrupted and the ability of MuSCs to repair injured muscle markedly declines. Until recently, this decline has been largely attributed to extrinsic age-related alterations in the microenvironment to which MuSCs are exposed. However, as highlighted in this Perspective, recent reports show that MuSCs also progressively undergo cell-intrinsic alterations that profoundly affect stem cell regenerative function with aging. A more comprehensive understanding of the interplay of stem cell-intrinsic and extrinsic factors will set the stage for improving cell therapies capable of restoring tissue homeostasis and enhancing muscle repair in the aged.
                Bookmark

                Author and article information

                Contributors
                svbrooks@umich.edu
                Journal
                Aging Cell
                Aging Cell
                10.1111/(ISSN)1474-9726
                ACEL
                Aging Cell
                John Wiley and Sons Inc. (Hoboken )
                1474-9718
                1474-9726
                02 June 2021
                June 2021
                : 20
                : 6 ( doiID: 10.1111/acel.v20.6 )
                : e13393
                Affiliations
                [ 1 ] Department of Molecular & Integrative Physiology University of Michigan Ann Arbor MI USA
                [ 2 ] Department of Biomedical Engineering University of Michigan Ann Arbor MI USA
                [ 3 ] Department of Pathology Lipidomics Core Facility Wayne State University Detroit MI USA
                Author notes
                [*] [* ] Correspondence

                Susan V. Brooks, Department of Molecular & Integrative Physiology, University of Michigan, 2029 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109‐2002, USA.

                Email: svbrooks@ 123456umich.edu

                Author information
                https://orcid.org/0000-0002-5348-1464
                Article
                ACEL13393
                10.1111/acel.13393
                8208786
                34075679
                a68b48e9-02fc-4916-aa52-774e96d1153f
                © 2021 The Authors. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 07 March 2021
                : 29 June 2020
                : 08 May 2021
                Page count
                Figures: 7, Tables: 1, Pages: 16, Words: 9533
                Funding
                Funded by: American Federation for Aging Research , open-funder-registry 10.13039/100000965;
                Funded by: Biomedical Engineering Department, University of Michigan , open-funder-registry 10.13039/100008557;
                Funded by: National Institute on Aging , open-funder-registry 10.13039/100000049;
                Award ID: 1R01AG050676‐01A1
                Award ID: 3P01AG051442‐03S1
                Award ID: 5P30AG024824‐10
                Funded by: Glenn Foundation for Medical Research , open-funder-registry 10.13039/100001642;
                Funded by: Department of Molecular & Integrative Physiology, University of Michigan
                Funded by: National Center for Research Resources , open-funder-registry 10.13039/100000097;
                Award ID: 1S10RR027926‐01
                Funded by: National Institute of Arthritis and Musculoskeletal and Skin Diseases , open-funder-registry 10.13039/100000069;
                Award ID: 1P30AR069620‐01
                Funded by: U.S. Department of Defense , open-funder-registry 10.13039/100000005;
                Award ID: W81XWH18SBAA1‐1257992
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                June 2021
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.0.2 mode:remove_FC converted:16.06.2021

                Cell biology
                aging,cellular immunology,inflammation,injury,mass spectrometry,sarcopenia,satellite stem cell,skeletal muscle

                Comments

                Comment on this article