10.6084/m9.figshare.5624836.v2 Bojana Kravic Bojana Kravic Angelika B. Harbauer Angelika B. Harbauer Vanina Romanello Vanina Romanello Luca Simeone Luca Simeone F.-Nora Vögtle F.-Nora Vögtle Tobias Kaiser Tobias Kaiser Marion Straubinger Marion Straubinger Danyil Huraskin Danyil Huraskin Martin Böttcher Martin Böttcher Cristina Cerqua Cristina Cerqua Eva Denise Martin Eva Denise Martin Daniel Poveda-Huertes Daniel Poveda-Huertes Andreas Buttgereit Andreas Buttgereit Adam J. Rabalski Adam J. Rabalski Dieter Heuss Dieter Heuss Rüdiger Rudolf Rüdiger Rudolf Oliver Friedrich Oliver Friedrich David Litchfield David Litchfield Michael Marber Michael Marber Leonardo Salviati Leonardo Salviati Dimitrios Mougiakakos Dimitrios Mougiakakos Winfried Neuhuber Winfried Neuhuber Marco Sandri Marco Sandri Chris Meisinger Chris Meisinger Said Hashemolhosseini Said Hashemolhosseini In mammalian skeletal muscle, phosphorylation of TOMM22 by protein kinase CSNK2/CK2 controls mitophagy Taylor & Francis Group 2018 CSNK2/CK2 CSNK2B homeostasis mitochondria mitophagy p62 PINK1 skeletal myopathy TOMM22 2018-02-01 15:04:55 Dataset https://tandf.figshare.com/articles/dataset/In_mammalian_skeletal_muscle_phosphorylation_of_TOMM22_by_protein_kinase_CSNK2_CK2_controls_mitophagy/5624836 <p>In yeast, Tom22, the central component of the TOMM (translocase of outer mitochondrial membrane) receptor complex, is responsible for the recognition and translocation of synthesized mitochondrial precursor proteins, and its protein kinase CK2-dependent phosphorylation is mandatory for TOMM complex biogenesis and proper mitochondrial protein import. In mammals, the biological function of protein kinase CSNK2/CK2 remains vastly elusive and it is unknown whether CSNK2-dependent phosphorylation of TOMM protein subunits has a similar role as that in yeast. To address this issue, we used a skeletal muscle-specific <i>Csnk2b</i>/<i>Ck2β</i>-conditional knockout (cKO) mouse model. Phenotypically, these skeletal muscle <i>Csnk2b</i> cKO mice showed reduced muscle strength and abnormal metabolic activity of mainly oxidative muscle fibers, which point towards mitochondrial dysfunction. Enzymatically, active muscle lysates from skeletal muscle <i>Csnk2b</i> cKO mice phosphorylate murine TOMM22, the mammalian ortholog of yeast Tom22, to a lower extent than lysates prepared from controls. Mechanistically, CSNK2-mediated phosphorylation of TOMM22 changes its binding affinity for mitochondrial precursor proteins. However, in contrast to yeast, mitochondrial protein import seems not to be affected in vitro using mitochondria isolated from muscles of skeletal muscle <i>Csnk2b</i> cKO mice. PINK1, a mitochondrial health sensor that undergoes constitutive import under physiological conditions, accumulates within skeletal muscle <i>Csnk2b</i> cKO fibers and labels abnormal mitochondria for removal by mitophagy as demonstrated by the appearance of mitochondria-containing autophagosomes through electron microscopy. Mitophagy can be normalized by either introduction of a phosphomimetic TOMM22 mutant in cultured myotubes, or by in vivo electroporation of phosphomimetic <i>Tomm22</i> into muscles of mice. Importantly, transfection of the phosphomimetic <i>Tomm22</i> mutant in muscle cells with ablated <i>Csnk2b</i> restored their oxygen consumption rate comparable to wild-type levels. In sum, our data show that mammalian CSNK2-dependent phosphorylation of TOMM22 is a critical switch for mitophagy and reveal CSNK2-dependent physiological implications on metabolism, muscle integrity and behavior.</p>