zum Inhalt springen

2023

December

Moschandrea C, Kondylis V, Evangelakos I, Herholz M, Schneide F, Schmidt C, Yang M, Ehret S, Heine H, Jaeckstein MJ, Szczepanowska K, Schwarzer R, Baumann L, Bock T, Nikitopoulou E, Brodesser S, Krüger M, Frezza C, Heeren J, Trifunovic A*, Pasparakis M* (2023). Mitochondrial dysfunction abrogates dietary lipid processing in enterocytes. Nature. DOI: 10.1038/s41586-023-06857-0. *corresponding authors

October

den Brave F, Schulte U, Fakler B, Pfanner N, Becker T (2023). Mitochondrial complexome and import network. Trends Cell Biol. S0962-8924(23)00208-8. DOI: 10.1016/j.tcb.2023.10.004.

Guhathakurta S, Erdogdu NU, Hoffmann JJ, Grzadzielewska I, Schendzielorz A, Seyfferth J, Mårtensson CU, Corrado M, Karoutas A, Warscheid B, Pfanner N, Becker T, Akhtar A (2023). COX17 acetylation via MOF-KANSL complex promotes mitochondrial integrity and function. Nat Metab. DOI: 10.1038/s42255-023-00904-w.

Schorn, F., Werthenbach, J. P., Hoffmann, M., Daoud, M., Stachelscheid, J., Schiffmann, L.M., Hildebrandt, X., Lyu, S.I., Peltzer, N., Quaas, A., Vucic, D., Silke, J., Pasparakis, M., Kashkar, H. (2023). cIAPs control RIPK1 kinase activity-dependent and -independent cell death and tissue inflammation. The EMBO Journal 2023 Oct 4:e113614. DOI: 10.15252/embj.2023113614

August

Anton, V., Buntenbroich, I., Simões, T., Joaquim, M., Müller, L., Büttner, R., Odenthal, M., Hoppe, T., Escobar-Henriques, M. (2023). E4 ubiquitin ligase promotes mitofusin turnover and mitochondrial stress response. Molecular Cell. DOI: doi.org/10.1016/j.molcel.2023.07.021. (Open access).

July

Czabotar, P.E., García-Sáez, A.J. (2023). Mechanisms of BCL-2 family proteins in mitochondrial apoptosis. Nat Rev Mol Cell Biol. 2023 Jul 12. doi: 10.1038/s41580-023-00629-4.

June

Buntenbroich, I., Anton, V., Perez-Hernandez, D., Simões, T., Gaedke, F., Schauss, A., Dittmar, G., Riemer, J., Escobar-Henriques, M. (2023).  Docking and stability defects in mitofusin highlight the proteasome as a potential therapeutic target. iScience. 2023 Jun 7;26(7):107014. doi: 10.1016/j.isci.2023.107014

May

Peker, E., Weiss, K., Song, J., Zarges, C., Gerlich, S., Boehm, V., Trifunovic, A., Langer, T., Gehring, N., Becker, T. and Riemer, J. (2023). A two-step mitochondrial import pathway couples the disulfide relay with matrix complex I biogenesis. J. Cell Biol. 222, e202210019.

Coyne, L.P., Wang, X., Song, J., de Jong, E., Schneider, K., Massa, P.T., Middleton, F.A., Becker, T. and Chen, X.J. (2023). Mitochondrial protein import clogging as a mechanism of disease. eLife 12, e84330.

April

Montoro-Gámez, C., Nolte, H., Molinié, T., Evangelista, G., Tröder, S., Barth, E., Popovic, M., Trifunovic, A., Zevnik, B., Langer, T., Rugarli E.I. SARM1 deletion delays cerebellar but not spinal cord degeneration in an enhanced mouse model of SPG7 deficiency. Brain. 2023 Apr 22:awad136. doi: 10.1093/brain/awad136. Online ahead of print. PMID: 37086482

March

Flores-Romero, H., Dadsena, S., García-Sáez, A.J. (2023). Mitochondrial pores at the crossroad between cell death and inflammatory signaling. Mol Cell. 2023 Mar 16;83(6):843-856. doi: 10.1016/j.molcel.2023.02.021.

Hevler, J. F., Albanese, P., Cabrera-Orefice, A., Potter, A., Jankevics, A., Misic, J., Scheltema, R.A., Brandt, U., Arnold, S.,  and Heck, A.J.R. (2023) MRPS36 provides a structural link in the eukaryotic 2-oxoglutarate dehydrogenase complex. Open Bio. 2023 Mar;13(3):220363. doi: 10.1098/rsob.220363. Epub 2023 Mar 1.

February

Li, Q. and Hoppe, T. (2023). Role of amino acid metabolism in mitochondrial homeostasis. Front Cell Dev Biol. 10.3389/fcell.2023.1127618

January

Schulte, U.; den Brave, F., Haupt, A., Gupta, A., Song, J., Müller, C.S., Engelke, J., Mishra, S., Mårtensson, C., Ellenrieder, L., Priesnitz, C., Straub, S.P., Doan, K.N., Kulawiak, B., Bildl, W., Rampelt, H., Wiedemann, N., Pfanner, N., Fakler, B. and Becker, T. Mitochondrial complexome reveals quality control pathways of protein import (2023). Nature. doi.org/10.1038/s41586-022-05641-w.

Takeda, H., Busto, J.V., Lindau, C., Tsutsumi, A., Tomii, K., Imai, K., Yamamori, Y., Hirokawa, T., Motono, C., Ganesan, I., Wenz, L.S., Becker, T., Kikkawa, M., Pfanner, N., Wiedemann, N. and Endo, T. (2023) A multipoint guidance mechanism for b-barrel folding on the SAM complex. Nat. Struct. Mol. Biol. doi.org/10.1038/s41594-022-00897-2.

Song, J., Steidle, L., Steymans, I., Singh, J., Sanner, A., Böttinger, L., Winter, D. and Becker, T. (2023). The mitochondrial Hsp70 controls the assembly of the F1FO-ATP synthase. Nat. Commun. 14, 39.

2022

November

Hermeling, J. CW., Herholz, M., Baumann, L., Cores, E.C., Zečić, A., Hoppe, T., Riemer, J., Trifunovic, A. (2022). Mitochondria-originated redox signalling regulates KLF-1 to promote longevity in Caenorhabditis elegans. Redox Biology 2022,  https://doi.org/10.1016/j.redox.2022.102533

October

Jacobs, L.J., Hoehne, M.N., Riemer, J. (2022). Measuring Intracellular H2O2 in Intact Human Cells Using the Genetically Encoded Fluorescent Sensor HyPer7. Bio-protocol 12 (20).

Kožich, V, Schwahn, B.C., Sokolová, J., Křížková, M., Ditroi, T., Krijt, J., Khalil, Y., Křížek, T., Vaculíková-Fantlová, T., Stibůrková, B., Mills, P., Clayton, P., Barvíková, K., Blessing, H., Sykut-Cegielska, J., Dionisi-Vici, C., Gasperini, S., García-Cazorla, Á., Haack, T.B., Honzík, T., Ješina, P., Kuster, A., Laugwitz, L., Martinelli, D., Porta, F., Santer, R., Schwarz, G., Nagy, P. (2022). Human ultrarare genetic disorders of sulfur metabolism demonstrate redundancies in H2S homeostasis. Redox Biol. 2022 Dec;58:102517. doi: 10.1016/j.redox.2022.102517. Epub 2022 Oct 18.PMID: 36306676. Free PMC article.

Rödl, S., den Brave, F., Räschle, M., Kizmaz, B., Lenhard, S., Groh, C., Becker, H., Zimmermann, J., Morgan, B., Richling, E., Becker, T. and Herrmann, J.M. (2022). The metabolite-controlled ubiquitin conjugase Ubc8 promotes mitochondrial protein import. Life Sci. Alliance 6, e202201526.

Bolgi, O., Silva-Garcia, M., Ross, B., Pilla, E., Kari, V., Killisch, M., Spitzner, M., Stark, N., Lenz, C., Weiss, K., Donzelli, L., Gorrell, M.D., Grade, M., Riemer, J., Urlaub, H., Dobbelstein, M., Huber, R., Geiss-Friedlander, R. (2022). Dipeptidyl peptidase 9 triggers BRCA2 degradation and promotes DNA damage repair. EMBO Rep. e54136.

September

Ravanelli, S., Li, Q., Annibal, A., Trifunovic, A., Antebi, A., Hoppe, T. (2022). Reprograming of proteasomal degradation by branched chain amino acid metabolism.  Aging Cell 2022, https://doi.org/10.1111/acel.13725

Ahola, M., Rivera, M. P., Hermans, S., Chandragiri, S., Giavalisco, P., Nolte, H., Langer, T. (2022). OMA1-mediated integrated stress response protects against ferroptosis in mitochondrial cardiomyopathy.  Cell Metab. 2022 Sep 8;S1550-4131(22)00360-6. doi: 10.1016/j.cmet.2022.08.017.

August

Jacobs, L.J., Riemer, J. (2022) Maintenance of small molecule redox homeostasis in mitochondria. FEBS Lett. doi: 10.1002/1873-3468.

Balaji, V., Müller, L., Lorenz, R., Kevei, E., Zhang, W.H., Santiago, U., Gebauer, J., Llamas, E., Vilchez, D., Camacho, C.J., Pokrzywa, W., Hoppe T. (2022). A Dimer-Monomer Switch Controls CHIP-Dependent Substrate Ubiquitylation and Processing. Mol Cell. 2022, doi: https://doi.org/10.1016/j.molcel.2022.08.003

Patron, M., Tarasenko, D., Nolte, H., Ghosh, M.,  Ohba, Y., Lasarzewski, Y., Ahmadi, Z.A., Cabrera-Orefice, A., Eyiama, A., Kellermann, T., Rugarli, E.I., Brandt, U., Meinecke, M., Langer, T. (2022). Regulation of mitochondrial proteostasis by the proton gradient. The EMBO Journal (2022)e110476. https://doi.org/10.15252/embj.2021110476

July

Salscheider, S.L., Gerlich, S., Cabrera-Orefice, A., Peker, E., Rothemann, R.A., Murschall, L.M., Finger, Y., Szczepanowska, K., Ahmadi, Z.A., Guerrero-Castillo, S., Erdogan, A., Becker, M., Ali, M., Habich, M., Petrungaro, C., Burdina, N., Schwarz, G., Klußmann, M., Neundorf, I., Stroud, D.A., Ryan, M.T., Trifunovic, A., Brandt, U., Riemer, J. (2022). AIFM1 is a component of the mitochondrial disulfide relay that drives complex I assembly through efficient import of NDUFS5. The EMBO Journal (2022)e110784. https://doi.org/10.15252/embj.2022110784

Flores-Romero, H., García-Sáez, A.J. (2022). MERLIN: A BRET-Based Proximity Biosensor for Studying Mitochondria-ER Contact Sites. Methods Mol Biol. 2022;2525:197-205. doi: 10.1007/978-1-0716-2473-9_14.

May

Schatton, D., Di Pietro, G., Szczepanowska, K., Veronese, M., Marx, M.-C., Braunöhler, K., Esther, B., Müller, S., Giavalisco, P., Langer, T., Trifunovic, A., Rugarli, I.E. (2022). CLUH controls astrin-1 expression to couple mitochondrial metabolism to cell cycle progression. Elife. 2022 May 13;11:e74552. DOI: 10.7554/eLife.74552

Hoffmann, J.J. and Becker, T. (2022) Crosstalk between mitochondrial protein import and lipids. Int. J. Mol. Sci. 23, 5275.

April

Croon, M., Szczepanowska, K., Popovic, M., Lienkamp, C., Senft, K., Brandscheid, C.P., Bock, T., Gnatzy-Feik, L., Ashurov, A., Acton, R.J., Kaul, H., Pujol, C., Rosenkranz, S., Krüger, M., Trifunovic, A. (2022). FGF21 modulates mitochondrial stress response in cardiomyocytes only under mild mitochondrial dysfunction. Sci Adv. 2022 Apr 8;8(14):eabn7105

Priesnitz, C., Böttinger, L., Zufall, N., Gebert, M., Guiard, B., van der Laan, M. and Becker, T. (2022) Coupling to Pam16 differentially controls the dual role of Pam18 in protein import and respiratory chain formation. Cell Rep. 39, 1108619.

March

Song, J. and Becker T. (2022). Fidelity of organellar protein targeting. Curr. Opin. Cell Biol. 75:102071.

Rumyantseva, A., Popovic, M., Trifunovic, A. (2022). CLPP deficiency ameliorates neurodegeneration caused by impaired mitochondrial protein synthesis. Brain (Oxford Academic) awab303, https://doi.org/10.1093/brain/awab303

Cosentino, K., Hertlein, V., Jenner, A., Dellmann, T., Gojkovic, M., Peña-Blanco, A., Dadsena, S., Wajngarten, N., Danial, J.S.H., Thevathasan, J.V., Mund, M., Ries, J., Garcia-Saez, A.J. (2022).
The interplay between BAX and BAK tunes apoptotic pore growth to control mitochondrial-DNA-mediated inflammation. Mol Cell. 2022 Mar 3;82(5):933-949.e9. doi: 10.1016/j.molcel.2022.01.008. Epub 2022 Feb 3.

February

Wani, G.A., Sprenger, H.-G., Ndoci, K., Chandragiri, S., Acton, R.J., Schatton, D., Kochan, S.M.V., Sakthivelu, V., Jevtic, M., Seeger, J.M., Müller, S., Giavalisco, P., Rugarli, E.I., Motori, E., Langer, T., Bergami, M. (2022). Metabolic control of adult neural stem cell self-renewal by the mitochondrial protease YME1L. Cell Reports • DOI:https://doi.org/10.1016/j.celrep.2022.110370

Hoehne, M.N., Jacobs, L.J.H.C., Lapacz, K.J., Calabrese, G., Murschall, L.M., Marker, T., Kaul, H., Trifunovic, A., Morgan, B., Fricker, M., Belousov, V.V., Riemer J. (2022). Spatial and temporal control of mitochondrial H2O2 release in intact human cells. EMBO J, e109169

Gibhardt, C.S., Riemer, J., Bogeski, I. (2022). Calcium and redox signals at mitochondrial interfaces: A nanoview perspective. Cell Calcium 103:102550

January

Li, X., Straub, J., Medeiros, T.C., Den Brave, F., Peker, E., Atanassov, I., Stillger, K., Michaelis, J.B., Burbridge, E., Adrain, C., Münch, C., Riemer, J., Becker, T., Pernas, L.F. (2022). Mitochondria shed their outer membrane in response to infection-induced stress. Science  • Vol 375, Issue 6577 • DOI: 10.1126/science.abi4343

 

2021

Flores-Romero, H., Hohorst, L., John, M., Albert, M-C., King, L. E, Beckmann, L., Szabo, T., Hertlein, V., Luo, X., Villunger, A., Frenzel, L. P., Kashkar, H., Garcia-Saez, A. J. (2021). BCL-2-family protein tBID can act as a BAX-like effector of apoptosis. The EMBO Journal (2021)e108690. https://doi.org/10.15252/embj.2021108690

Willenborg, S., Sanin, D.E., Jais, A., Ding, X., Ulas, T., Nüchel, J., Popović, M., MacVicar, T., Langer, T., Schultze, J.L., Gerbaulet, A., Roers, A., Pearce, E.J., Brüning, J.C., Trifunovic, A., Eming, S.A. (2021). Mitochondrial metabolism coordinates stage-specific repair processes in macrophages during wound healing.  Cell Metabolism (2021), https://doi.org/10.1016/j.cmet.2021.10.004

Winter, D. and Becker, T. (2021). Surveying the mitochondrial proteome. Nature Cell Biology 23, 1216-1217.

den Brave, F., Gupta, A., Becker, T. (2021) Protein Quality Control at the Mitochondrial Surface. Front. Cell Dev. Biol. DOI: https://doi.org/10.3389/fcell.2021.795685

Kaczmarek, A.T., Bender, D., Gehling, T., Kohl, J.B., Daimagüler, H.S., Santamaria-Araujo, J.A., Liebau, M.C., Koy, A., Cirak, S., Schwarz, G.J. (2021). A defect in molybdenum cofactor binding causes an attenuated form of sulfiteoxidase deficiency. Inherit Metab Dis. 2022 Mar;45(2):169-182. Do i: 10.1002/jimd.12454. Epub 2021 Nov 24.PMID: 34741542

Guerrero-Castillo, S., van Strien J., Brandt, U. and Arnold, S. (2021) Ablation of mitochondrial DNA results in widespread remodeling of the mitochondrial complexome. EMBO Journal, 40, e108648.

Kaczmarek, A.T., Bahlmann, N., Thaqi, B., May, P., Schwarz, G. (2021). Machine learning-based identification and characterization of 15 novel pathogenic SUOX missense mutations. 2021 Sep-Oct;134(1-2):188-194. doi: 10.1016/j.ymgme.2021.07.011. Epub 2021 Aug 8.PMID: 34420858

Hevler J.F., Chiozzi, R.Z., Cabrera-Orefice, A., Brandt U., Arnold, S, and Albert J. R. Heck, A.J.R. (2021) Molecular characterization of a complex of apoptosis-inducing factor 1 with cytochrome c oxidase of the mitochondrial respiratory chain. Proc. Natl. Acad. Sci. USA, 118, e2106950118.

den Brave, F., Engelke, J. and Becker, T. (2021) Quality control of protein import into mitochondria. Biochem. J. 478, 3125-3143.

Bock, T., Türk, C., Aravamudhan, S., Keufgens, L., Bloch, W., Rozsivalova, D.H., Romanello, V., Nogara, L., Blaauw, B., Trifunovic, A., Braun, T., Krüger, M. (2021). PERM1 interacts with the MICOS-MIB complex to connect the mitochondria and sarcolemma via ankyrin B. Nat Commun. 2021 Aug 12;12(1):4900

Geueke A., Mantellato G., Kuester F., Schettina P., Nelles M., Seeger J.M., Kashkar H., Niemann C. (2021)
The anti-apoptotic Bcl-2 protein regulates hair follicle stem cell function
EMBO Rep (2021)e52301. https://doi.org/10.15252/embr.202052301

Murschall, L,M*., Peker, E*., MacVicar, T., Langer, T., Riemer, J. (2021). Protein Import Assay into Mitochondria Isolated from Human Cells. Bio-protocol 11 (12), e4057-e4057

Peker, E., Erdogan, A.J., Volkov, A.N., Riemer, J. (2021). Erv1 and Cytochrome c Mediate Rapid Electron Transfer via A Collision-Type Interaction. J Mol Biol. 433(15):167045

Schlagowski, A.M., Knöringer, K., Morlot, S., Sánchez Vicente, A., Flohr, T., Krämer, L., Boos, F., Khalid, N., Ahmed, S., Schramm, J., Murschall, L.M., Haberkant, P., Stein, F., Riemer, J., Westermann, B., Braun, R.J., Winklhofer, K.F., Charvin, G., Herrmann, J.M. (2021). Increased levels of mitochondrial import factor Mia40 prevent the aggregation of polyQ proteins in the cytosol. EMBO J. e107913

Kaspar S., Oertlin C., Szczepanowska K., Kukat A., Senft K., Lucas C., Brodesser S., Hatzoglou M., Larsson O., Topisirovic I., Trifunovic A. (2021)
Adaptation to mitochondrial stress requires CHOP-directed tuning of ISR
Science Advances  26 May 2021: Vol. 7, no. 22, eabf0971; DOI: 10.1126/sciadv.abf0971

Szczepanowska, K., Trifunovic, A. (2021). "Mitochondrial matrix proteases: quality control and beyond. " FEBS J. 2021 May 10

Müller, L., Kutzner, C.E., Balaji, V., Hoppe, T. (2021). In vitro analysis of E3 ubiquitin ligase function. J Vis Exp. 171, doi: 10.3791/62393.

He L., Sehrawat T.S., Verma V.K, Navarro-Corcuera A., Sidhu G., Mauer A., Luo X., Katsumi T., Chen J., Shah S., Arab J.P., Cao S., Kashkar H., Gores G.J., Malhi H., Shah V.H. (2021)
XIAP Knockdown in Alcohol-Associated Liver Disease Models Exhibits Divergent in vitro and in vivo Phenotypes Owing to a Potential Zonal Inhibitory Role of SMAC
Front. Physiol., 07 May 2021 

Aravamudhan, S., Türk, C., Bock, T., Keufgens, L., Nolte, H., Lang, F., Krishnan, R.K., König, T., Hammerschmidt, P., Schindler, N., Brodesser, S., Rozsivalova, D.H., Rugarli, E., Trifunovic, A., Brüning, J., Langer, T., Braun, T., Krüger, M. (2021). Phosphoproteomics of the developing heart identifies PERM1 - An outer mitochondrial membrane protein.  2021 Feb 5;154:41-59. doi.org/10.1016/j.yjmcc.2021.01.010

Kelly, B., Carrizo, G.E., Edwards-Hicks, J., Sanin, D.E., Stanczak, M.A., Priesnitz, C., Flachsmann, L.J., Curtis, J.D., Mittler, G., Musa, Y., Becker, T., Buescher, J.M. and Pearce, E.L. (2021) Sulfur sequestration promotes multicellularity during nutrient limitation. Nature 591, 471-476.

Takeda, H., Tsutsumi, A., Nishizawa, T., Lindau C., Busto, J.V., Wenz, L.S., Ellenrieder, L., Imai, K., Straub, S.P., Mossmann, W., Qiu, J., Yamamori, Y., Tomii, K., Suzuki, J., Murata, T., Ogasawara, S., Nureki, O., Becker, T., Pfanner, N., Wiedemann, N., Kikkawa, M. and Endo, T. (2021) Mitochondrial sorting and assembly machinery operates by b-barrel switching. Nature 590, 163-169.

Song, J., Herrmann, J.M. and Becker, T. (2021) Quality control of the mitochondrial proteome. Nat. Rev. Mol. Cell Biol. 22, 54-70.

Mellis, A.-T., Roeper, J., Misko, A.L., Kohl, J., Schwarz, G. (2021). Sulfite Alters the Mitochondrial Network in Molybdenum Cofactor Deficiency.  doi: 10.3389/fgene.2020.594828.

Mellis, A.-T., Misko, A.L., Arjune, S., Liang, Y., Erdélyi, K., Ditrói, T., Kaczmarek, A. T., Nagy, P., Schwarz, G. (2021).
The role of glutamate oxaloacetate transaminases in sulfite biosynthesis and H 2 S metabolism. Redox Biol. 2021 Jan;38:101800. doi: 10.1016/j.redox.2020.101800.

Gupta, A. and Becker, T. (2021) Mechanisms and pathways of mitochondrial outer membrane protein biogenesis. Biochim. Biophys. Acta 1862, 148323.

2020

den Brave, F. and Becker, T. (2020) Supercomplex formation boosts respiration. EMBO Rep. e51830

Bonekamp, N.A., Peter, B., Hillen, H.S. Felser, A., Bergbrede, T., Choidas, A., Horn, M., Unger, A., Di Lucrezia, R., Atanassov,, I., Li, X., Koch, U., Menninger, S., Boros,, J., Habenberger, P., Giavalisco, P., Cramer, P., Denzel, M.S., Nussbaumer, P., Klebl, B., Falkenberg, M., Gustafsson, C.M., Larsson, N.G  (2020). Small-molecule inhibitors of human mitochondrial DNA transcription. Nature 588, 712–716. https://doi.org/10.1038/s41586-020-03048-z

Rumyantseva, A., Motori, E., Trufunovic, A. (2020). DARS2 is indispensable for Purkinje cell survival and protects against cerebellar ataxia". Hum Mol Genet.; 2020 Oct 10;29(17):2845-2854.     
DOI: 10.1093/hmg/ddaa176.

Kirschberg M, Heuser S, Marcuzzi GP, Hufbauer M, Seeger JM, Đukić A, Tomaić V, Majewski S, Wagner S, Wittekindt C, Würdemann N, Klussmann JP, Quaas A, Kashkar H, Akgül B. (2020)
ATP synthase modulation leads to an increase of spare respiratory capacity in HPV associated cancers.
Sci Rep. 2020 Oct 15;10(1):17339. doi: 10.1038/s41598-020-74311-6. PMID: 33060693

Bergami M., Motori E., (2020) Reweaving the Fabric of Mitochondrial Contact Sites in Astrocytes. DOI: 10.3389/fcell.2020.592651.

Albert, M.C., Brinkmann, K., Pokrzywa, W., Günther, S.D., Krönke, M., Hoppe, T*., Kashkar, H*. (2020). CHIP ubiquitylates NOXA and induces its lysosomal degradation in response to DNA damage. Cell Death Dis. 11:740 *Corresponding author

Joaquim M., Escobar-Henriques M. (2020). Role of Mitofusins and Mitophagy in Life or Death Decisions. DOI: 10.3389/fcell.2020.572182

Escobar-Henriques M., Vincent A. (2020) Mitochondrial Surveillance by Cdc48/p97: MAD vs. Membrane Fusion. DOI: 10.3390/ijms21186841

Motori, E., Atanassov, I., Kochan, S.M.V., Folz-Donahue, K., Sakthivelu, V., Giavalisco, P., Toni, N., Puyal, J., Larsson, N.-G. (2020). Neuronal metabolic rewiring promotes resilience to neurodegeneration caused by mitochondrial dysfunctionScience Advances; Vol. 6, no. 35, eaba8271; DOI: 10.1126/sciadv.aba8271

Finger, Y., Habich, M., Gerlich, S., Urbanczyk, S., Logt, E., Koch, J., Schu, L., Lapacz, K.J., Ali, M., Petrungaro, C., Salscheider, S.L., Pichlo, C., Baumann U., Mielenz, D., Brachvogel, B., Hofmann, K., Riemer, J. (2020). Proteasomal degradation induced by DPP9‐mediated processing competes with mitochondrial protein import. EMBO Journal 2020 Aug e103889

Göbel, J., Engelhardt, E., Pelzer, P., Sakthivelu, V., Jahn, H.M., Jevtic, M., Folz-Donahue, K., Kukat, C., Schauss, A., Frese, C.K., Giavalisco, P., Ghanem, A., Conzelmann, K-K., Motori, E., Bergami, M. (2020). Mitochondria-Endoplasmic Reticulum Contacts in Reactive Astrocytes Promote Vascular Remodeling. Cell Metab. 2020 Apr 7; 31(4): 791–808.e8.; https://doi.org/10.1016/j.cmet.2020.03.005

Schiffmann, L.M., Werthenbach, J. P., Heintges-Kleinhofer, F., Seeger, J.M., Fritsch, M., Günther, S. D. Willenborg, S., Brodesser, S., Lucas, C., Jüngst, C., Albert, M. C., Schorn, F., Witt, A., Moraes, C. T., Bruns, C. J., Pasparakis, M., Krönke, M., Eming, S. A., Coutelle, O., and Kashkar, H. (2020) . Mitochondrial respiration controls neoangiogenesis during wound healing and tumour growth. Nat. Commun. 11, 3653 (2020). https://doi.org/10.1038/s41467-020-17472-2. University of Cologne, Press release

Hofsetz, E., Demir, F., Szczepanowska, K., Kukat, A., Kizhakkedathu[1], J., Trifunovic, A., Huesgen, P.F. (2020). The Mouse Heart Mitochondria N Terminome Provides Insights Into ClpXP-mediated Proteolysis. Mol Cell Proteomics ;mcp.RA120.002082. DOI: 10.1074/mcp.RA120.002082. Online ahead of print.

Murschall, L.M., Gerhards, A., MacVicar, T., Peker, E., Hasberg, L., Wawra, S., Langer, T., Riemer, J. (2020). The C-terminal region of the oxidoreductase MIA40 stabilizes its cytosolic precursor during mitochondrial import.
DOI: 10.1186/s12915-020-00824-1

Hoppe, T., Cohen, E. (2020). Organismal Protein Homeostasis Mechanisms. DOI: 10.1534/genetics.120.301283

Timper, K., del Río-Martín, A., Cremer, A.L., Bremser, S., Alber, J. Giavalisco, P. Varela, L., Heilinger, C., Nolte, H., Trifunovic, A., Horvath, T.L., Kloppenburg, P., Backes, H., Brüning, J.C.  (2020). GLP-1 Receptor Signaling in Astrocytes Regulates Fatty Acid Oxidation, Mitochondrial Integrity, and Function. Cell Metab. S1550-4131(20)30240-0. doi: 10.1016/j.cmet.2020.05.001. Online ahead of print.

Finger, Y., Riemer, J. (2020). Protein import by the mitochondrial disulfide relay in higher eukaryotes. Biol Chem. 2020 May 26;401(6-7):749-763

Ravanelli, S., den Brave, F., Hoppe, T. (2020). Mitochondrial Quality Control Governed by Ubiquitin. Front. Cell Dev. Biol. | doi: 10.3389/fcell.2020.00270

Szczepanowska, K., Senft, K., Heidler, J., Herholz, M., Kukat, M.A., Höhne, N., Hofsetz, E., Becker, C.,  Kaspar, S., Giese, H., Zwicker, K., Guerrero-Castillo, S, Baumann, L., Kauppila, J., Rumyantseva, A., Müller, S., Frese, C.K., Brandt, U., Riemer, J., Wittig, I., and Trifunovic, A. (2020) A salvage pathway maintains highly functional respiratory complex I. Nature Communications 11, 1643.

Pla-Martin, D.,  Schatton, D., Wiederstein, J.L., Marx, M.C.,  Khiati, S., Krüger, M., Rugarli, E.I. (2020). CLUH granules coordinate translation of mitochondrial proteins with mTORC1 signaling and mitophagy. EMBO J. e102731. doi.org/10.15252/embj.2019102731. [Epub ahead of print]

Deshwal, S., Fiedler, K.U., and Langer, T. (2020). Mitochondrial Proteases: Multifaceted Regulators of Mitochondrial Plasticity. Annu. Rev. Biochem.,  doi: 10.1146/annurev-biochem-062917-012739. [Epub ahead of print]

Bender, D., Kaczmarek, A.T., Kuester, S., Burlina, A.B., and Schwarz G. (2020). Oxygen and nitrite reduction by heme-deficient sulphite oxidase in a patient with mild sulphite oxidase deficiency. J. Inherit. Metab. Dis. Jan 17; doi: 10.1002/jimd.12216. [Epub ahead of print]

Mayr, S.J., Röper, J., and Schwarz, G. (2020). Alternative splicing of the bicistronic gene molybdenum cofactor synthesis 1 (MOCS1) uncovers a novel mitochondrial protein maturation mechanism. J. Biol. Chem., Jan 29. pii: jbc.RA119.010720. doi: 10.1074/jbc.RA119.010720. [Epub ahead of print]

Balaji, V. and Hoppe, T. (2020). Regulation of E3 ubiquitin ligases by homotypic and heterotypic assembly.  F1000 Research 2020, 9 (F1000 Faculty Rev), 88.

Ricke, K.M., Paß, T., Kimoloi, S., Fährmann, K., Jüngst, C., Schauss, A., Baris, O., Aradjanski, M.,  Trifunovic, A., Eriksson Faelker, T., Bergami, M. and Wiesner R. (2020). Mitochondrial dysfunction combined with high calcium load leads to impaired antioxidant defense underlying the selective loss of nigral dopaminergic neurons. J. Neurosci., 26;40(9):1975-1986.

Schütter, M. and Graef, M. (2020). Localized de novo phospholipid synthesis drives autophagosome biogenesis. Autophagy 1-2. doi: 10.1080/15548627.2020.1725379. [Epub ahead of print]

Ricke, K.M., Paß, T., Kimoloi, S., Fährmann, K., Jüngst, C., Schauss, A., Baris, O.R., Aradjanski, M., Trifunovic, A., Eriksson Faelker, T.M., Bergami, M., Wiesner, R.J. (2020). Mitochondrial Dysfunction Combined with High Calcium Load Leads to Impaired Antioxidant Defense Underlying the Selective Loss of Nigral Dopaminergic Neurons. J Neurosci. 2020 Feb 26;40(9):1975-1986

Schütter M, Giavalisco P, Brodesser S, and Graef, M. (2020). Local fatty acid channeling into phospholipid synthesis drives phagophore expansion during autophagy.  Cell 180, 135-149.e14. doi: 10.1016/j.cell.2019.12.005. Epub 2019 Dec 26.

2019

MacVicar, T., Ohba, Y., Nolte, H., Mayer, F., Tatsuta, T., Sprenger, H.G., Lindner, B., Zhao, Y., Li, J., Bruns, C., Krüger, M., Habich, M., Riemer, J., Schwarzer, R., Pasparakis, M., Henschke, S., Brüning, J.C., Zamboni, N. and Langer, T. (2019). Lipid signalling drives proteolytic rewiring of mitochondria by YME1L. Nature Nature 575 , 361-365.

Murru, S., Hess, S., Barth, E., Almajan, E.R., Schatton, D., Hermans, S., Brodesser, S., Langer, T., Kloppenburg, P., Rugarli, E.I. (2019). Astrocyte-specific deletion of the mitochondrial m-AAA protease reveals glial contribution to neurodegeneration. GLIA 67, 1526-1541.

Sprenger, H.G. and Langer, T. (2019). The good and the bad of mitochondrial breakups. Trends Cell Biol. 29, 888-900. doi: 10.1016/j.tcb.2019.08.003. [Epub ahead of print].

Richter, F., Dennerlein, S., Nikolov, M., Jans, D., Naumenko, N., Aich, A., MacVicar, T., Linden, A., Jakobs, S., Urlaub, H., Langer, T. and Rehling, P. (2019). ROMO1 is a constituent of the human presequence translocase required for YME1L protease import. J. Cell Biol. 218, 598-614.

Schuster, R., Anton, V., Simoes, T., Altin, S., den Brave, F., Hermanns, T., Hospenthal, M., Komander, D., Dittmar, G., Dohmen, R.J., Escobar-Henriques, M. (2019). Dual role of a GTPase conformational switch for membrane fusion by mitofusin ubiquitylation. Life Sci. Alliance, 3(1). pii: e201900476. doi: 10.26508/lsa.201900476. Print 2020 Jan.

Anton, V., Buntenbroich, I., Schuster, R., Babatz, F., Simoes, T., Altin, S., Calabrese, G., Riemer, J., Schauss, A.C., Escobar-Henriques, M. (2019). Plasticity in salt-bridge allows fusion-competent ubiquitylation of mitofusins and Cdc48 recognition. Life Sci. Alliance, 2(6). pii: e201900491.

Escobar-Henriques, M. and Joaquim, M. (2019). Mitofusins: Disease Gatekeepers and Hubs in Mitochondrial Quality Control by E3 Ligases. Front. Physiol. 10, 517.

Escobar-Henriques, M., Altin, S., and Brave, F.D. (2019). Interplay between the Ubiquitin Proteasome System and Mitochondria for Protein Homeostasis. (Book chapter), in: Sumoylation and Ubiquitylation: Current and Emerging Concepts, Caister Academic Publishing. Open access in: Curr. Issues Mol. Biol. 35, 35-58.

Gross, A. and Graef, M. (2019). Mechanisms of autophagy in metabolic stress response. J. Mol. Biol., pii: S0022-2836(19)30559-5. DOI: 10.1016/j.jmb.2019.09.005. [Epub ahead of print].

Cui, R., Medeiros, T.C., Willemsen, D., Iasi, L.N.M., Collier, G.E., Graef, M., Reichard, M., and Valenzano, D.R. (2019). Relaxed selection limits lifespan by increasing mutation load. Cell 178, 385-399.

Silva Ramos, E., Motori, E., Brüser, C., Kühl, I., Yeroslaviz, A., Ruzzenente, B., Kauppila, J.H.K., Busch, J.D., Hultenby, K., Habermann, B.H., Jakobs, S., Larsson, N.G., and Mourier, A. (2019). Mitochondrial fusion is required for regulation of mitochondrial DNA replication. PLoS Genet. 15, e1008085.

Rangaraju, V.*, Lewis, T.*, Hirabayashi, Y.*, Bergami, M.*, Motori, E.*, Cartoni, R.*, Kwon, S.K., and Courchet, J. (2019). Pleiotropic Mitochondria: The influence of mitochondria on neuronal development and disease. J. Neurosci. 39, 8002-8208. *equal contribution

Ren, S.Q., Li, Z., Lin, S., Bergami, M., and Shi, S.H. (2019). Precise long-range microcircuit-to-microcircuit communication connects the frontal and sensory cortices in the mammalian brain. Neuron. 104, 385-401.e3.

Sprenger, H.G., Wani, G., Hesseling, A., König, T., Patron, M., MacVicar, T., Ahola, S., Wai, T., Barth, E., Rugarli, E.I., Bergami, M., Langer, T. (2019). Loss of the mitochondrial i‐AAA protease YME1L leads to ocular dysfunction and spinal axonopathy. EMBO Mol. Med. 11 pii: e9288.

Göbel, J., Pelzer, P., Engelhardt, E., Sakthivelu, V., Jahn, H.M., Milica, J., Folz-Donahue, K., Schauss, A., Ghanem, A., Conzelmann,, K.K., Frese, C., Motori, E., Bergami, M. (2019). Mitochondrial fusion in reactive astrocytes coordinates local metabolic domains to promote vascular repair. BioRxiv, doi: https://doi.org/10.1101/657999.

Hammerschmidt, P., Ostkotte, D., Nolte, H., Gerl, M.J., Jais, A., Brunner, H.L., Sprenger, H.G., Awazawa, M., Nicholls, H.T., Turpin-Nolan, S.M., Langer, T., Krüger, M., Brügger, B., and Brüning, J.C. (2019). CerS6-derived sphingolipids interact with Mff and promote mitochondrial fragmentation in obesity. Cell 177, 1536-1552.

Raichur, S., Brunner, B., Bielohuby, M., Hansen, G., Pfenninger, A., Wang, B., Brüning, J.C., Larsen, P.J., and Tennagels, N. (2019). The role of C16:0 ceramide in the development of obesity and type 2 diabetes: CerS6 inhibition as a novel therapeutic approach. Mol. Metab. 21, 36-50.

Turpin-Nolan, S.M., Hammerschmidt, P., Chen, W., Jais, A., Timper, K., Awazawa, M., Brodesser, S., and Brüning, J.C. (2019). CerS1-Derived C18:0 Ceramide in Skeletal Muscle Promotes Obesity-Induced Insulin Resistance. Cell Rep. 26, 1-10.

Herholz, M., Cepeda, E., Baumann, L., Kukat, A., Hermeling, J., Maciej, S., Szczepanowska, K., Pavlenko, V., Frommolt, P., and Trifunovic, A.. (2019). KLF-1 orchestrates a xenobiotic detoxification program essential for longevity of mitochondrial mutants. Nat. Commun. 10, 3323. DOI: 10.1038/s41467-019-11275-w. 

Calabrese, G., Peker, E., Amponsah, P.S., Hoehne, M.N., Riemer, T., Mai, M., Bienert, G.P., Deponte, M., Morgan, B., and Riemer, J. (2019). Hyperoxidation of mitochondrial peroxiredoxin limits H2O2-induced cell death in yeast. EMBO J. e101552. doi.org/10.15252/embj.2019101552. [Epub ahead of print]

Habich, M., Salscheider, S.L., Murschall, L.M., Hoehne, M.N., Fischer, M., Schorn, F., Petrungaro, C., Ali, M., Erdogan, A.J., Abou-Eid, S., Kashkar, H., Dengjel, J., and Riemer, J. (2019). Vectorial import via a metastable disulfide-linked complex allows for a quality control step and import by the mitochondrial disulfide relay. Cell Rep. 26, 759-774.

Gräb, J., Suarez, I., van Gumpel, E., Winter, S., Schreiber, F., Esser, A., Holscher, C., Fritsch, M., Herb, M., Schramm, M., Wachsmuth, L., Pallasch, C., Pasparakis, M., Kashkar, H., and Rybniker, J. (2019). Corticosteroids inhibit Mycobacterium tuberculosis-induced necrotic host cell death by abrogating mitochondrial membrane permeability transition. Nat. Commun. 10, 688.

Fritsch, M., Günther, S.D., Schwarzer, R., Albert, M.C., Schorn, F., Werthenbach, J.P., Schiffmann, L.M., Stair, N., Stocks, H., Seeger, J.M., Lamkanfi, M., Krönke, M., Pasparakis, M., and Kashkar, H. (2019). Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature 575, 683-687.

Günther, S.D., Fritsch, M., Seeger, J.M., Schiffmann, L.M., Snipas, S.J., Coutelle, M., Kufer, T.A., Higgins, P.G., Hornung, V., Bernardini, M.L., Höning, S., Krönke, M., Salvesen, G.S., and Kashkar, H. (2019). Cytosolic Gram-negative bacteria prevent apoptosis by inhibition of effector caspases through LPS. Nat. Microbiol., Epub ahead of print. DOI 10.1038/s41564-019-0620-5.

Holzer, T., Probst, K., Etich, J., Auler, M., Georgieva, V., Bluhm, B., Frie, C., Heilig, J., Niehoff, A., Nüchel, J., Plomann, M., Seeger, J.M., Kashkar, H., Baris, O.R., Wiesner, R.J., and Brachvogel, B. (2019). Respiratory chain inactivation links cartilage-mediated growth retardation to mitochondrial diseases. J. Cell Biol. 218, 1853-1870

Schiffmann, L.M., Fritsch, M., Gebauer, F., Günther, S.D., Stair, N.R., Seeger, J.M., Thangarajah, F., Dieplinger, G., Bludau, M., Alakus, H., Göbel, H., Quaas, A., Zander, T., Hilberg, F., Bruns, C.J., Kashkar, H., and Coutelle, O. (2019). Tumour-infiltrating neutrophils counteract anti-VEGF therapy in metastatic colorectal cancer. Br. J. Cancer 120, 69-78.

Benkert, J., Hess, S., Roy, S., Beccano-Kelly, D., Wiederspohn, N., Duda, J., Simons, C., Patil, K., Gaiffulina, A., Mannal, N., Dragicevic, E., Spaich, D., Müller, S., Nemeth, J., Hollmann, H., Deuter, N., Mousba, Y, Kubisch, C., Poetschke, C., Striessnig, J., Pongs, O., Schneider, T., Wade-Martins, R., Patel, S., Parlato, R., Frank, T., Kloppenburg, P., and Liss, B. (2019). Cav2.3 channels contribute to dopaminergic neuron loss in a model of Parkinson’s disease. Nat. Commun. 10, 5094.

Bugiardini, E., Mitchell, A.L., Rosa, I.D., Horning-Do, H.T., Pitmann, A., Poole, O.V., Holton, J.L., Shah, S., Woodward, C., Hargreaves, I., Quinlivan, R., Amunts, A., Wiesner, R.J., Houlden, H., Holt, I.J., Hanna, M.G., Pitceathly, R.D.S., Spinazzola, A. (2019). MRPS25 mutations impair mitochondrial translation and cause encephalomyopathy. Hum. Mol. Genet. 19 Apr 30. pii: ddz093. doi: 10.1093/hmg/ddz093. [Epub ahead of print]

Murru, S., Hess, S., Barth, E., Almajan, E.R., Schatton, D., Hermans, S., Brodesser, S., Langer, T., Kloppenburg, P., Rugarli, E.I. (2019). Astrocyte-specific deletion of the mitochondrial m-AAA protease reveals glial contribution to neurodegeneration. GLIA 67, 1526-1541.

Lubomirov, L.T., Gagov, H., Schroeter, M.M., Wiesner, R.J., and Franko, A. (2019). Augmented contractility of murine femoral arteries in a streptozotocin diabetes model is related to increased phosphorylation of MYPT1. Physiol Rep. 7(3):e13975. doi: 10.14814/phy2.13975.

Bender, D., Kaczmarek, A.T., Santamaria-Araujo, J.A., Stueve, B., Waltz, S., Bartsch, D., Kurian, L., Cirak, S., and Schwarz, G. (2019). Impaired mitochondrial maturation of sulfite oxidase in a patient with severe sulfite oxidase deficiency. Hum. Mol. Genet. 28, 2885-2899.

Bender, D., Kaczmarek, AT., Niks, D., Hille, R., and Schwarz, G. (2019). Mechanism of nitrite-dependent NO synthesis by human sulfite oxidase. Biochem. J. 476, 1805-1815.

Sriramachandran, A.M., Meyer-Teschendorf, K., Pabst, S., Ulrich, H.D., Gehring, N.H., Hofmann, K., Praefcke, G.J.K. and Dohmen, R.J. (2019). Arkadia/RNF111 is a SUMO-targeted ubiquitin ligase with preference for substrates marked with SUMO1-capped SUMO2/3 chain. Nat. Comm. doi.org/10.1038/s41467-019-11549-3. [Epub ahead of print]

Kohl, J.B., Mellis, A.T., and Schwarz, G. (2019). Homeostatic impact of sulfite and hydrogen sulfide on cysteine catabolism. Br. J. Pharmacol. 176, 554-570.

Kaczmarek, A.T., Strampraad, M.J.F., Hagedoorn, P.L., and Schwarz, G. (2019). Reciprocal regulation of sulfite oxidation and nitrite reduction by mitochondrial sulfite oxidase. Nitric Oxide. 89, 22-31.

Zivanovic, J., Kouroussis, E., Kohl, J.B., ... Schwarz, G., Snyder, S.H., Paul, B.D., Carroll, K.S., Filipovic, M.R. (2019). Selective Persulfide Detection Reveals Evolutionarily Conserved Antiaging Effects of S-Sulfhydration. Cell Metab. 30, 1152-1170.e13.

Pabst, S., Döring, L.M., Petreska, N., Dohmen. R.J. (2019). Methods to study SUMO dynamics in yeast. Methods Enzymol. 618, 187-210.

Anton, V., Buntenbroich, I., Schuster, R., Babatz, F., Simoes, T., Altin, S., Calabrese, G., Riemer, J., Schauss, A.C., Escobar-Henriques, M. (2019). Plasticity in salt-bridge allows fusion-competent ubiquitylation of mitofusins and Cdc48 recognition. Life Sci. Alliance, 2(6). pii: e201900491.

2018

Sprenger, H.G., Wani, G., Hesseling, A., König, T., Patron, M., MacVicar, T., Ahola, S., Wai, T., Barth, E., Rugarli, E.I., Bergami, M., Langer, T. (2018). Loss of the mitochondrial i‐AAA protease YME1L leads to ocular dysfunction and spinal axonopathy. EMBO Mol Med. pii: e9288. doi: 10.15252/emmm.201809288. [Epub ahead of print]

Schuster, R., Simoes, T., den Brave, F., and Escobar-Henriques, M. (2018). Separation and visualization of low abundant ubiquitylated forms. BIO-Protocols 8, DOI: 10.21769/BioProtoc.3081.

Simões, T., Schuster, R., den Brave, F., Escobar-Henriques, M. (2018). Cdc48 regulates a deubiquitylase cascade critical for mitochondrial fusion. Elife 7. pii: e30015. doi: 10.7554/eLife.30015. [Epub ahead of print].

Medeiros, T.C., Thomas, R.L., Ghillebert, R., and Graef, M. (2018). Autophagy balances mtDNA synthesis and degradation by DNA polymerase POLG during starvation. J. Cell Biol. 217, 1601-1611.

Medeiros, T.C., and Graef, M. (2018). Autophagy determines mtDNA copy number dynamics during starvation. Autophagy 15, 178-179.

Graef, M. (2018). Lipid droplet-mediated lipid and protein homeostasis in budding yeast. FEBS letters 592, 1291-1303.

Schatton, D. and Rugarli, E.I. (2018). A concert of RNA-binding proteins coordinates mitochondrial function. Crit. Rev. Biochem. Mol. Biol. 53, 652-666.

Schatton, D., and Rugarli, E.I. (2018). Post-transcriptional regulation of mitochondrial function. Curr. Opin. Physiol. 3, 6-15.

Bonekamp, N.A., and Larsson, N.G. (2018). SnapShot: Mitochondrial Nucleoid. Cell 172, 388-388.e1.

Göbel, J. Motori, E., and Bergami, M. (2018). Spatiotemporal control of mitochondrial network dynamics in astroglial cells. Biochem. Biophys. Res. Commun. 500, 17-25.

Jahn, H.M., Bergami, M. (2018). Critical periods regulating the circuit integration of adult-born hippocampal neurons. Cell Tissue Res. 371, 23-32.

Timper, K., Paeger, L., Sánchez-Lasheras, C., Varela, L., Jais, A., Nolte, H., Vogt, M.C., Hausen, A.C., Heilinger, C., Evers, N., Pospisilik, J.A., Penninger, J.M., Taylor, E.B., Horvath, T.L., Kloppenburg, P., Brüning, J.C.. (2018). Mild impairment of mitochondrial OXPHOS promotes fatty acid utilization in POMC neurons and improves glucose homeostasis in obesity. Cell Rep. 25, 383-397.

Brandt, C., Nolte, H., Henschke, S., Engström, Ruud, L., Awazawa, M., Morgan, D.A., Gabel, P., Sprenger, H.G., Hess, M.E., Günther, S., Langer, T., Rahmouni, K., Fenselau, H., Krüger, M., and Brüning, J.C. (2018). Food Perception Primes Hepatic ER-Homeostasis via Melanocortin-Dependent Control of mTOR-Activation. Cell Nov 15;175, 1321-1335.

Becker, C., Kukat, A., Szczepanowska, K., Hermans, S., Senft, K., Brandscheid, C.P., Maiti, P., and Trifunovic, A. (2018). CLPP deficiency protects against metabolic syndrome but hinders adaptive thermogenesis. EMBO Rep. 19, e45126.

Roma, L.P., Deponte, M., Riemer, J., and Morgan, B. (2018). Mechanisms and applications of redox-sensitive green fluorescent protein-based hydrogen peroxide probes. Antioxid. Redox Signaling 29, 552-568.

Habich, M., Salscheider, S.L., and Riemer, J. (2018). Cysteine residues in mitochondrial intermembrane space proteins: more than just import. Br. J. Pharmacol. 176, 514-531.

Erdogan, A. J., Ali, M., Habich, M., Salscheider, S. L., Schu, L., Petrungaro, C., Thomas, L. W., Ashcroft, M., Leichert, L. I., Roma, L. P., and Riemer, J. (2018). The mitochondrial oxidoreductase CHCHD4 is present in a semi-oxidized state in vivo. Redox Biol. 17, 200-206.

Mattie, S., Riemer, J., Wideman, J.G., McBride, H.M. (2018). A new mitofusin topology places the redox-regulated C terminus in the mitochondrial intermembrane space. J Cell Biol. 217, 507-515.

Meyer, A.J., Riemer, J., and Rouhier, N. (2018). Oxidative protein folding: state-of-the-art and current avenues of research in plants. New Phytol. 221, 1230-1246.

Chauhan, D., Bartok, E., Gaidt, M.M., Bock, F.J., Herrmann, J., Seeger, J.M., Broz, P., Beckmann, R., Kashkar, H., Tait, S.W.G., Müller, R., and Hornung, V. (2018). BAX/BAK-Induced Apoptosis Results in Caspase-8-Dependent IL-1β Maturation in Macrophages. Cell Rep. 25, 2354-2368.

Quiñones, M., Al-Massadi, O., Folgueira, C., Bremser, S., Gallego, R., Torres-Leal, L., Haddad-Tóvolli, R., García-Caceres, C., Hernandez-Bautista, R., Lam, B.Y.H., Beiroa, D., Sanchez-Rebordelo, E., Senra, A., Malagon, J.A., Valerio, P., Fondevila, M.F., Fernø, J., Malagon, M.M., Contreras, R., Pfluger, P., Brüning, J.C., Yeo, G., Tschöp, M., Diéguez, C, López, M., Claret, M., Kloppenburg, P., Sabio, G., and Nogueiras, R. (2018). p53 in AgRP neurons is required for protection against diet-induced obesity via JNK1. Nat. Commun. 9, 3432.

Weiland, D., Brachvogel, B., Hornig-Do, H.-T., Neuhaus, J.F.G., Holzer, T., Tobin, D.J., Niessen, C.M., Wiesner, R.J. and Baris, O.R. (2018). Imbalance of mitochondrial respiratory chain complexes in the epidermis induces severe skin inflammation. J. Invest Dermatol. 138, 132-140. #corresponding author

Bender, D., and Schwarz, G. (2018). Nitrite-dependent nitric oxide synthesis by molybdenum enzymes. FEBS Lett. 592, 2126-2139.

Nolte, H., MacVicar, T.D., Tellkamp, F., Krüger, M. (2018). Instant Clue: A Software Suite for Interactive Data Visualization and Analysis. Sci Rep. 8, 12648.

Wiederstein, J.L., Nolte, H., Günther, S., Piller, T., Baraldo, M., Kostin, S., Bloch, W., Schindler, N., Sandri, M., Blaauw, B., Braun, T., Hölper, S., Krüger, M. (2018). Skeletal Muscle-Specific Methyltransferase METTL21C Trimethylates p97 and Regulates Autophagy-Associated Protein Breakdown. Cell Rep. 23, 1342-1356.

2017

Jiang, M., Kauppila, T.E.S., Motori, E., Li, X., Atanassov, I., Folz-Donahue, K., Bonekamp, N.A., Albarran-Gutierrez, S., Stewart, J.B., and Larsson, N.G. (2017). Increased total mtDNA copy number cures male infertility despite unaltered mtDNA mutation load. Cell Metab. 26, 429-436.e4.

Erdogan, A. J. and Riemer, J. (2017). Mitochondrial disulfide relay and its substrates: mechanisms in health and disease. Cell Tissue Res. 367, 59-72.

Friederich, M.W., Erdogan, A.J., Coughlin, C.R., Elos, M.T., Jiang, H., O'Rourke, C.P., Lovell, M.A., Wartchow, E., Gowan, K., Chatfield, K.C., Chick, W.S., Spector, E.B., Van Hove, J.L.K., and Riemer, J. (2017). Mutations in the accessory subunit NDUFB10 result in isolated complex I deficiency and illustrate the critical role of intermembrane space import for complex I holoenzyme assembly. Hum. Mol. Genet. 26, 702-716.

Calabrese, G. Morgan, B., and Riemer, J. (2017). Mitochondrial Glutathione: Regulation and Functions. Antioxid. Redox Signaling 27, 1162-1177.

Habich, M. and Riemer, J. (2017). Detection of Cysteine Redox States in Mitochondrial Proteins in Intact Mammalian Cells. In: Mokranjac, D., Perocchi, F. (eds). Mitochondria. Methods in Molecular Biology, Vol 1567. Humana Press, New York, NY.

Döring, K., Ahmed, N., Riemer, T., Suresh, H.G., Vainshtein, Y., Habich, M., Riemer, J., Mayer, M.P., O'Brien, E.P., Kramer, G., and Bukau B. (2017). Profiling Ssb-Nascent Chain Interactions Reveals Principles of Hsp70-Assisted Folding. Cell. 170, 298-311.

Tawo, R., Pokrzywa, W., Kevei, E., Akyuz, M.E., Balaji, V., Arian, S., Höhfeld, J., and Hoppe, T. (2017). The Ubiquitin Ligase CHIP Integrates Proteostasis and Aging by Regulation of Insulin Receptor Turnover. Cell 169, 470-482.

Saita, S., Nolte, H., Fiedler, K.U., Kashkar, H., Venne, A.S., Zahedi, R.P., Krüger, M., and Langer, T. (2017). PARL mediates Smac proteolytic maturation in mitochondria to promote apoptosis. Nat Cell Biol. 19, 318-328.

Schiffmann, L.M., Brunold, M., Liwschitz, M., Goede, V., Loges, S., Wroblewski, M., Quaas, A., Alakus, H., Stippel, D., Bruns, C.J., Hallek, M., Kashkar, H., Hacker, U.T., Coutelle, O. (2017). A combination of low-dose bevacizumab and imatinib enhances vascular normalisation without inducing extracellular matrix deposition. Br. J. Cancer. 116, 600-608.

Paeger, L., Pippow, A., Hess, S., Paehler, M., Klein, A.C., Husch, A., Pouzat, C., Brüning, J.C., Kloppenburg, P. (2017). Energy imbalance alters Ca2+ handling and excitability of POMC neurons. Elife, 6. pii: e25641. doi: 10.7554/eLife.25641.

Ortner, N.J., Bock, G., Dougalis, A., Kharitonova, M., Duda, J., Hess, S., Tuluc P., Pomberger, T., Stefanova, N., Pitterl, F., Ciossek, T., Oberacher, H., Draheim, H.J., Kloppenburg, P., Liss, B., Striessnig, J. (2017). Lower affinity of isradipine for L-Type Ca2+ channels during substantia nigra dopamine neuron-like activity: implications for neuroprotection in Parkinson’s disease. J. Neurosci. 37, 6761-6777.

Schommers, P., Thurau, A., Bultmann-Mellin, I., Guschlbauer, M., Klatt, A.R., Rozman, J., Klingenspor, M., Hrabe de Angelis, M., Alber, J., Gründemann, D., Sterner-Kock, A., and Wiesner, R.J. (2017). Metformin causes a futile intestinal–hepatic cycle which increases energy expenditure and slows down development of a type 2 diabetes-like state. Mol. Metabolism. 6, 737-747.

Neuhaus, J.F.G., Baris, Kittelmann, O. R. A. Becker, K. Rothschild, M.A., and Wiesner, R.J. (2017). Catecholamine metabolism induces mitochondrial DNA deletions and leads to severe adrenal degeneration during aging. Neuroendocrinology 104, 72-84.

Stöckigt, F., Beiert, T., Knappe, V., Baris, O.R., Wiesner, R.J., Clemen, C.S., Nickenig, G., Andrié, R.P. and Schrickel, J.W. (2017): Aging-related mitochondrial dysfunction facilitates the occurrence of serious arrhythmia after myocardial infarction. Biochem. Biophys. Res. Comm. 493, 604-610.

Kumar, A. Dejanovic, B., Hetsch, F., Semtner, M., Fusca, D., Arjune, S., Santamaria-Araujo, J.A., Winkelmann, A., Ayton, S., Bush, A.I., Kloppenburg, P., Meier, J.C., Schwarz, G., and Belaidi, A.A. (2017). S-sulfocysteine/NMDA receptor-dependent signaling underlies neurodegeneration in molybdenum cofactor deficiency. J. Clin. Invest. 127, 4365-4378.

Schatton, D., Pla-Martin, D., Marx, M.C., Hansen, H., Mourier, A., Nemazanyy, I., Pessia, A., Zentis, P., Corona, T., Kondylis, V., Barth, E., Schauss, A.C., Velagapudi, V., and Rugarli, E.I. (2017). CLUH regulates mitochondrial metabolism by controlling translation and decay of target mRNAs. J. Cell Biol. 216, 675-693. doi: 10.1083/jcb.201607019.

2016

MacVicar, T. and Langer, T. (2016). OPA1 processing in cell death and disease: the long and short of it. J. Cell Sci. 129, 2297-2306.

Korwitz, A., Merkwirth, C., Richter-Dennerlein, R., Tröder, S.E., Sprenger, H.-G., Qui­ros, P.M., López-Otín, C., Rugarli, E.I., and Langer, T. (2016). Loss of OMA1 delays neurodegeneration by preventing stress-induced OPA1 processing in mitochondria. J. Cell Biol. 212, 157-166.

Wang, S., Jacquemyn, J., Murru, S., Martinelli, P., Barth, E., Langer, T., Niessen, C.M., and Rugarli, E.I. (2016). The mitochondrial m-AAA protease prevents demyelination and hair greying. PLoS Genet 12, e1006463.

Velázquez, A.P., Tatsuta, T., Ghillebert, R., Drescher, I., and Graef, M. (2016). Lipid droplet-mediated ER homeostasis regulates autophagy and cell survival during starvation. J. Cell Biol. 212, 621-631.

Velazquez, A.P., and Graef, M. (2016). Autophagy regulation depends on ER homeostasis controlled by lipid droplets. Autophagy 12, 1409-1410.

Seiferling, D., Szczepanowska, K., Becker, C., Senft, K., Hermans, S., Maiti, P., König, T., Kukat, A., Trifunovic, A. (2016). Loss of CLPP alleviates mitochondrial cardiomyopathy without affecting the mammalian UPRmt. EMBO Rep. 17, 953-964.

Guarás, A., Perales-Clemente, E., Calvo, E., Acín-Pérez, R., Loureiro-Lopez, M., Pujol, C., Martínez-Carrascoso, I., Nuñez, E., García-Marqués, F., Rodríguez-Hernández, M.A., Cortés, A., Diaz, F., Pérez-Martos, A., Moraes, C.T., Fernández-Silva, P., Trifunovic, A., Navas, P., Vazquez, J., Enríquez, J.A. (2016). The CoQH2/CoQ ratio serves as a sensor of respiratory chain efficiency. Cell Rep. 15, 197-209.

Franz, A., Ackermann, L. and Hoppe, T.  (2016). Ring of change: CDC48/p97 drives protein dynamics at chromatin. Front. Genet. 7, 73.

Vlantis, K., Wullaert, A., Polykratis, A., Kondylis, V., Dannappel, M., Schwarzer, R., Welz, P., Corona, T., Walczak, H., Weih, F., Klein, U., Kelliher, M., and Pasparakis, M. (2016). NEMO Prevents RIP Kinase 1-Mediated Epithelial Cell Death and Chronic Intestinal Inflammation by NF-kappaB-Dependent and -Independent Functions. Immunity 44, 553-567.

Knittel, G., Liedgens, P., Korovkina, D., Seeger, J. M., Al-Baldawi, Y., Al-Maarri, M., Fritz, C., Vlantis, K., Bezhanova, S., Scheel, A. H., Wolz, O. O., Reimann, M., Möller, P., López, C., Schlesner, M., Lohneis, P., Weber, A. N., Trümper, L., German International Cancer Genome Consortium Molecular Mechanisms in Malignant Lymphoma by Sequencing Project Consortium, Staudt, L. M., Ortmann, M., Pasparakis, M., Siebert, R., Schmitt, C. A., Klatt, A. R., Wunderlich, F. T., Schäfer, S. C., Persigehl, T., Montesinos-Rongen, M., Odenthal, M., Büttner, R., Frenzel, L. P., Kashkar, H., Reinhardt, H. C. (2016). B-cell-specific conditional expression of Myd88p.L252P leads to the development of diffuse large B-cell lymphoma in mice. Blood 127, 2732-41.

Lehtonen, J. M., Forsstrom, S., Bottani, E., Viscomi, C., Baris, O. R., Isoniemi, H., Hockerstedt, K., Osterlund, P., Hurme, M., Jylhava, J., Leppa, S., Markkula, R., Helio, T., Mombelli, G., Uusimaa, J., Laaksonen, R., Laaksovirta, H., Auranen, M., Zeviani, M., Smeitink, J., Wiesner, R. J., Nakada, K., Isohanni, P., Suomalainen, A. (2016). FGF21 is a biomarker for mitochondrial translation and mtDNA maintenance disorders. Neurology 87, 2290-2299.

Szczepanowska, K., Maiti, P., Kukat, A., Hofsetz, E., Nolte, H., Senft, K., Becker, C., Ruzzenente, B., Hornig-Do, H.-T., Wibom, R., Wiesner, R.J., Krüger, M. and Trifunovic, A. (2016). CLPP coordinates mitoribosomal assembly through regulation of ERAL1 levels. EMBO J. 35, 2566-2583.

Franko, A., Huypens, P., Neschen, S., Irmler, M., Rozman J., Rathkolb, B., Neff, F., Prehn, C., Dubois, G., Baumann, M., Massinger, R., Gradinger, D., Przemeck, G.K., Repp, B., Aichler, M., Feuchtinger, A., Schommers, P., Stöhr, O., Sanchez-Lasheras, C., Adamski, J., Peter, A., Prokisch, H., Beckers, J., Walch, A.K., Fuchs, H., Wolf, E., Schubert, M., Wiesner, R.J., and Hrabě de Angelis (2016). Bezafibrate improves insulin sensitivity and metabolic flexibility in STZ treated diabetic mice. Diabetes 65, 2540-2552.

 

 

 

zum Seitenanfang