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

      Mitochondrial dysfunction in macrophages promotes inflammation and suppresses repair after myocardial infarction

      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

          Innate immune cells play important roles in tissue injury and repair following acute myocardial infarction (MI). Although reprogramming of macrophage metabolism has been observed during inflammation and resolution phases, the mechanistic link to macrophage phenotype is not fully understood. In this study, we found that myeloid-specific deletion (mKO) of mitochondrial complex I protein, encoded by Ndufs4, reproduced the proinflammatory metabolic profile in macrophages and exaggerated the response to LPS. Moreover, mKO mice showed increased mortality, poor scar formation, and worsened cardiac function 30 days after MI. We observed a greater inflammatory response in mKO mice on day 1 followed by increased cell death of infiltrating macrophages and blunted transition to the reparative phase during post-MI days 3–7. Efferocytosis was impaired in mKO macrophages, leading to lower expression of antiinflammatory cytokines and tissue repair factors, which suppressed the proliferation and activation of myofibroblasts in the infarcted area. Mitochondria-targeted ROS scavenging rescued these impairments, improved myofibroblast function in vivo, and reduced post-MI mortality in mKO mice. Together these results reveal a critical role of mitochondria in inflammation resolution and tissue repair via modulation of efferocytosis and crosstalk with fibroblasts. These findings have potential significance for post-MI recovery as well as for other inflammatory conditions.

          Abstract

          Related collections

          Most cited references44

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

          Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes.

          Mononuclear phagocytes are versatile cells that can express different functional programs in response to microenvironmental signals. Fully polarized M1 and M2 (or alternatively activated) macrophages are the extremes of a continuum of functional states. Macrophages that infiltrate tumor tissues are driven by tumor-derived and T cell-derived cytokines to acquire a polarized M2 phenotype. These functionally polarized cells, and similarly oriented or immature dendritic cells present in tumors, have a key role in subversion of adaptive immunity and in inflammatory circuits that promote tumor growth and progression.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Conditional gene targeting in macrophages and granulocytes using LysMcre mice.

            Conditional mutagenesis in mice has recently been made possible through the combination of gene targeting techniques and site-directed mutagenesis, using the bacteriophage P1-derived Cre/loxP recombination system. The versatility of this approach depends on the availability of mouse mutants in which the recombinase Cre is expressed in the appropriate cell lineages or tissues. Here we report the generation of mice that express Cre in myeloid cells due to targeted insertion of the cre cDNA into their endogenous M lysozyme locus. In double mutant mice harboring both the LysMcre allele and one of two different loxP-flanked target genes tested, a deletion efficiency of 83-98% was determined in mature macrophages and near 100% in granulocytes. Partial deletion (16%) could be detected in CD11c+ splenic dendritic cells which are closely related to the monocyte/macrophage lineage. In contrast, no significant deletion was observed in tail DNA or purified T and B cells. Taken together, LysMcre mice allow for both specific and highly efficient Cre-mediated deletion of loxP-flanked target genes in myeloid cells.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              TLR signaling augments macrophage bactericidal activity through mitochondrial ROS

              Reactive oxygen species (ROS) are essential components of the innate immune response against intracellular bacteria, and it is thought that professional phagocytes generate ROS primarily via the phagosomal NADPH oxidase (Phox) machinery 1 . However, recent studies have suggested that mitochondrial ROS (mROS) also contribute to macrophage bactericidal activity, although the mechanisms linking innate immune signaling to mitochondria for mROS generation remain unclear 2-4 . Here we demonstrate that engagement of a subset of Toll-like receptors (TLR1, TLR2 and TLR4) results in the recruitment of mitochondria to macrophage phagosomes and augments mROS production. This response involves translocation of the TLR signaling adapter tumor necrosis factor receptor-associated factor 6 (TRAF6) to mitochondria where it engages evolutionarily conserved signaling intermediate in Toll pathways (ECSIT), a protein implicated in mitochondrial respiratory chain assembly 5 . Interaction with TRAF6 leads to ECSIT ubiquitination and enrichment at the mitochondrial periphery, resulting in increased mitochondrial and cellular ROS generation. ECSIT and TRAF6 depleted macrophages exhibit decreased levels of TLR-induced ROS and are significantly impaired in their ability to kill intracellular bacteria. Additionally, reducing macrophage mROS by expressing catalase in mitochondria results in defective bacterial killing, confirming the role of mROS in bactericidal activity. These results therefore reveal a novel pathway linking innate immune signaling to mitochondria, implicate mROS as important components of antibacterial responses, and further establish mitochondria as hubs for innate immune signaling.
                Bookmark

                Author and article information

                Contributors
                Journal
                J Clin Invest
                J Clin Invest
                J Clin Invest
                The Journal of Clinical Investigation
                American Society for Clinical Investigation
                0021-9738
                1558-8238
                15 February 2023
                15 February 2023
                15 February 2023
                : 133
                : 4
                : e159498
                Affiliations
                [1 ]Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
                [2 ]Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA.
                [3 ]Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA.
                [4 ]Medical Scientist Training Program, University of Washington School of Medicine, Seattle, Washington, USA.
                Author notes
                Address correspondence to: Rong Tian, Mitochondria and Metabolism Center, University of Washington, Seattle, Washington 98109, USA. Email: rongtian@ 123456uw.edu . MZ’s present address is: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.

                Authorship note: SC and MZ contributed equally to this work.

                Author information
                http://orcid.org/0000-0002-3753-0545
                http://orcid.org/0000-0003-1844-7081
                http://orcid.org/0000-0002-0930-6175
                http://orcid.org/0000-0003-4247-6755
                http://orcid.org/0000-0001-7181-9636
                http://orcid.org/0000-0003-2380-1321
                http://orcid.org/0000-0002-3676-3830
                Article
                159498
                10.1172/JCI159498
                9927948
                36480284
                3498d2c8-3ad1-4d4e-b0cd-0efe26dab4f5
                © 2023 Cai et al.

                This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 18 February 2022
                : 1 December 2022
                Funding
                Funded by: NIH
                Award ID: HL149695
                Funded by: NIH
                Award ID: HL133336
                Funded by: NIH
                Award ID: HL141187
                Award ID: HL142624
                Funded by: NIH
                Award ID: U19AI135976
                Funded by: American Heart Association, https://doi.org/10.13039/100000968;
                Award ID: 827259
                Funded by: the Japan Heart Foundation and the Uehara Memorial Foundation
                Award ID: 202040002
                to RT
                to SC
                to JD
                to GO
                to AY
                to AY
                Categories
                Research Article

                cardiology,metabolism,cardiovascular disease,macrophages,mitochondria

                Comments

                Comment on this article