Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Technical Report
  • Published:

Cell-type-specific profiling of brain mitochondria reveals functional and molecular diversity

Abstract

Mitochondria vary in morphology and function in different tissues; however, little is known about their molecular diversity among cell types. Here we engineered MitoTag mice, which express a Cre recombinase-dependent green fluorescent protein targeted to the outer mitochondrial membrane, and developed an isolation approach to profile tagged mitochondria from defined cell types. We determined the mitochondrial proteome of the three major cerebellar cell types (Purkinje cells, granule cells and astrocytes) and identified hundreds of mitochondrial proteins that are differentially regulated. Thus, we provide markers of cell-type-specific mitochondria for the healthy and diseased mouse and human central nervous systems, including in amyotrophic lateral sclerosis and Alzheimer’s disease. Based on proteomic predictions, we demonstrate that astrocytic mitochondria metabolize long-chain fatty acids more efficiently than neuronal mitochondria. We also characterize cell-type differences in mitochondrial calcium buffering via the mitochondrial calcium uniporter (Mcu) and identify regulator of microtubule dynamics protein 3 (Rmdn3) as a determinant of endoplasmic reticulum–mitochondria proximity in Purkinje cells. Our approach enables exploring mitochondrial diversity in many in vivo contexts.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: MitoTag mice allow isolation of intact and functional mitochondria from cells in situ.
Fig. 2: Immunocapture from MitoTag mice enriches cell-type-specific mitochondria without mitochondrial cross-contamination from other cells.
Fig. 3: Proteomic profiling of cell-type-specific mitochondria in the adult mouse cerebellum.
Fig. 4: Cell-type-specific mitochondrial markers are conserved within the CNS and across species, and they enable mitochondrial studies under pathological conditions.
Fig. 5: Astrocytic mitochondria metabolize long-chain fatty acids more efficiently than neurons.
Fig. 6: Granule cell mitochondria robustly buffer calcium via Mcu in contrast with Purkinje cell mitochondria.
Fig. 7: Rmdn3 mediates close ER–mitochondria juxtapositions in Purkinje cells.

Similar content being viewed by others

Data availability

All proteomics data generated within this study are deposited to the ProteomeXchange Consortium via the PRIDE44 partner repository with the dataset identifiers PXD010772, PXD010774, PXD010781 and PXD013380. In addition, Supplementary Data provide information on the proteomics data analysis used for the conclusions in the main and supplementary figures. All additional data that support the findings of this study are available from the corresponding author upon reasonable request. The MitoTag mouse line is available from The Jackson Laboratory as JAX#032675 (Rosa26–CAG–LSL–GFP–OMM). The McuFL/FL and the Rbp4:Cre mouse strains are protected under a material transfer agreement with the MMRRC (C57BL/6N-Mcutm1a(EUCOMM)Hmgu/H; Tg(Rbp4-cre)KL100Gsat/Mmucd), and the Rmdn3−/− mouse strain is protected under a material transfer agreement with the KOMP Repository (KO mouse project 3U01HG004080).

References

  1. MacIver, N. J., Michalek, R. D. & Rathmell, J. C. Metabolic regulation of T lymphocytes. Annu. Rev. Immunol. 31, 259–283 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Chen, H. & Chan, D. C. Mitochondrial dynamics in regulating the unique phenotypes of cancer and stem cells. Cell Metab. 26, 39–48 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Misgeld, T. & Schwarz, T. L. Mitostasis in neurons: maintaining mitochondria in an extended cellular architecture. Neuron 96, 651–666 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Devine, M. J. & Kittler, J. T. Mitochondria at the neuronal presynapse in health and disease. Nat. Rev. Neurosci. 19, 63–80 (2018).

    CAS  PubMed  Google Scholar 

  5. Nunnari, J. & Suomalainen, A. Mitochondria: in sickness and in health. Cell 148, 1145–1159 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. West, A. P., Shadel, G. S. & Ghosh, S. Mitochondria in innate immune responses. Nat. Rev. Immunol. 11, 389–402 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Pagliarini, D. J. et al. A mitochondrial protein compendium elucidates complex I disease biology. Cell 134, 112–123 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Shigeoka, T. et al. Dynamic axonal translation in developing and mature visual circuits. Cell 166, 181–192 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Ghazalpour, A. et al. Comparative analysis of proteome and transcriptome variation in mouse. PLoS Genet. 7, e1001393 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Heiman, M. et al. A translational profiling approach for the molecular characterization of CNS cell types. Cell 135, 738–748 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, W. W., Freinkman, E., Wang, T., Birsoy, K. & Sabatini, D. M. Absolute quantification of matrix metabolites reveals the dynamics of mitochondrial metabolism. Cell 166, 1324–1337.e1311 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Abu-Remaileh, M. et al. Lysosomal metabolomics reveals V-ATPase- and mTOR-dependent regulation of amino acid efflux from lysosomes. Science 358, 807–813 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ahier, A. et al. Affinity purification of cell-specific mitochondria from whole animals resolves patterns of genetic mosaicism. Nat. Cell Biol. 20, 352–360 (2018).

    CAS  PubMed  Google Scholar 

  14. Roh, H. C. et al. Simultaneous transcriptional and epigenomic profiling from specific cell types within heterogeneous tissues in vivo. Cell Rep. 18, 1048–1061 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hornig-Do, H. T. et al. Isolation of functional pure mitochondria by superparamagnetic microbeads. Anal. Biochem. 389, 1–5 (2009).

    CAS  PubMed  Google Scholar 

  16. Meeusen, S., McCaffery, J. M. & Nunnari, J. Mitochondrial fusion intermediates revealed in vitro. Science 305, 1747–1752 (2004).

    CAS  PubMed  Google Scholar 

  17. Sharma, K. et al. Cell type- and brain region-resolved mouse brain proteome. Nat. Neurosci. 18, 1819–1831 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Calvo, S. E., Clauser, K. R. & Mootha, V. K. MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins. Nucl. Acids Res. 44, D1251–D1257 (2016).

    CAS  PubMed  Google Scholar 

  19. Kang, J. S. et al. Docking of axonal mitochondria by syntaphilin controls their mobility and affects short-term facilitation. Cell 132, 137–148 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Sato, K., Yoshida, S., Fujiwara, K., Tada, K. & Tohyama, M. Glycine cleavage system in astrocytes. Brain Res. 567, 64–70 (1991).

    CAS  PubMed  Google Scholar 

  21. Miyake, S. et al. Identification and characterization of a novel mitochondrial tricarboxylate carrier. Biochem. Biophys. Res. Commun. 295, 463–468 (2002).

    CAS  PubMed  Google Scholar 

  22. Fowler, J. S. et al. Monoamine oxidase: radiotracer development and human studies. Methods 27, 263–277 (2002).

    CAS  PubMed  Google Scholar 

  23. Petrova, V. Y., Drescher, D., Kujumdzieva, A. V. & Schmitt, M. J. Dual targeting of yeast catalase a to peroxisomes and mitochondria. Biochem. J. 380, 393–400 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Fan, J., Li, X., Issop, L., Culty, M. & Papadopoulos, V. ACBD2/ECI2-mediated peroxisome-mitochondria interactions in Leydig cell steroid biosynthesis. Mol. Endocrinol. 30, 763–782 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Shai, N. et al. Systematic mapping of contact sites reveals tethers and a function for the peroxisome-mitochondria contact. Nat. Commun. 9, 1761 (2018).

    PubMed  PubMed Central  Google Scholar 

  26. Sinha, S., Bheemsetty, V. A. & Inamdar, M. S. A double helical motif in OCIAD2 is essential for its localization, interactions and STAT3 activation. Sci. Rep. 8, 7362 (2018).

    PubMed  PubMed Central  Google Scholar 

  27. Baughman, J. M. et al. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature 476, 341–345 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. De Stefani, D., Raffaello, A., Teardo, E., Szabò, I. & Rizzuto, R. A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter. Nature 476, 336–340 (2011).

    PubMed  PubMed Central  Google Scholar 

  29. De Vos, K. J. et al. VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum. Mol. Genet. 21, 1299–1311 (2012).

    PubMed  Google Scholar 

  30. Galmes, R. et al. ORP5/ORP8 localize to endoplasmic reticulum-mitochondria contacts and are involved in mitochondrial function. EMBO Rep. 17, 800–810 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Di Giorgio, F. P., Boulting, G. L., Bobrowicz, S. & Eggan, K. C. Human embryonic stem cell-derived motor neurons are sensitive to the toxic effect of glial cells carrying an ALS-causing mutation. Cell Stem Cell 3, 637–648 (2008).

    PubMed  Google Scholar 

  32. Schönfeld, P. & Reiser, G. Why does brain metabolism not favor burning of fatty acids to provide energy? Reflections on disadvantages of the use of free fatty acids as fuel for brain. J. Cereb. Blood Flow. Metab. 33, 1493–1499 (2013).

    PubMed  PubMed Central  Google Scholar 

  33. Ebert, D., Haller, R. G. & Walton, M. E. Energy contribution of octanoate to intact rat brain metabolism measured by 13C nuclear magnetic resonance spectroscopy. J. Neurosci. 23, 5928–5935 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Knobloch, M. et al. A fatty acid oxidation-dependent metabolic shift regulates adult neural stem cell activity. Cell Rep. 20, 2144–2155 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Eraso-Pichot, A. et al. GSEA of mouse and human mitochondriomes reveals fatty acid oxidation in astrocytes. Glia 66, 1724–1735 (2018).

    PubMed  Google Scholar 

  36. Harrington, J. L. & Murphy, E. The mitochondrial calcium uniporter: mice can live and die without it. J. Mol. Cell. Cardiol. 78, 46–53 (2015).

    CAS  PubMed  Google Scholar 

  37. Paillard, M. et al. Tissue-Specific mitochondrial decoding of cytoplasmic Ca(2+) signals is controlled by the stoichiometry of MICU1/2 and MCU. Cell Rep. 18, 2291–2300 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Eisenberg-Bord, M., Shai, N., Schuldiner, M. & Bohnert, M. A tether is a tether is a tether: tethering at membrane contact sites. Dev. Cell 39, 395–409 (2016).

    CAS  PubMed  Google Scholar 

  39. Lv, B. F. et al. Protein tyrosine phosphatase interacting protein 51 (PTPIP51) is a novel mitochondria protein with an N-terminal mitochondrial targeting sequence and induces apoptosis. Apoptosis 11, 1489–1501 (2006).

    CAS  PubMed  Google Scholar 

  40. Rangaraju, V., Lauterbach, M. & Schuman, E. M. Spatially stable mitochondrial compartments fuel local translation during plasticity. Cell 176, 73–84.e15 (2019).

    CAS  PubMed  Google Scholar 

  41. Lewis, T. L. Jr., Kwon, S. K., Lee, A., Shaw, R. & Polleux, F. MFF-dependent mitochondrial fission regulates presynaptic release and axon branching by limiting axonal mitochondria size. Nat. Commun. 9, 5008 (2018).

    PubMed  PubMed Central  Google Scholar 

  42. Alvarez-Castelao, B. et al. Cell-type-specific metabolic labeling of nascent proteomes in vivo. Nat. Biotechnol. 35, 1196–1201 (2017).

    CAS  PubMed  Google Scholar 

  43. Chen, C. L. et al. Proteomic mapping in live drosophila tissues using an engineered ascorbate peroxidase. Proc. Natl Acad. Sci. USA 112, 12093–12098 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Bayraktar, E. C. et al. MITO-Tag mice enable rapid isolation and multimodal profiling of mitochondria from specific cell types in vivo. Proc. Natl Acad. Sci. USA 116, 303–312 (2019).

    CAS  PubMed  Google Scholar 

  45. Kerschensteiner, M., Reuter, M. S., Lichtman, J. W. & Misgeld, T. Ex vivo imaging of motor axon dynamics in murine triangularis sterni explants. Nat. Protoc. 3, 1645–1653 (2008).

    PubMed  PubMed Central  Google Scholar 

  46. Sugiura, A., Mattie, S., Prudent, J. & McBride, H. M. Newly born peroxisomes are a hybrid of mitochondrial and ER-derived pre-peroxisomes. Nature 542, 251–254 (2017).

    CAS  PubMed  Google Scholar 

  47. Meera, P., Pulst, S. M. & Otis, T. S. Cellular and circuit mechanisms underlying spinocerebellar ataxias. J. Physiol. 594, 4653–4660 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Zaglia, T. et al. Content of mitochondrial calcium uniporter (MCU) in cardiomyocytes is regulated by microRNA-1 in physiologic and pathologic hypertrophy. Proc. Natl Acad. Sci. USA 114, E9006–E9015 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Dong, Z. et al. Mitochondrial Ca2+ uniporter is a mitochondrial luminal redox sensor that augments MCU channel activity. Mol. Cell 65, 1014–1028.e1017 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Henzi, T. & Schwaller, B. Antagonistic regulation of parvalbumin expression and mitochondrial calcium handling capacity in renal epithelial cells. PLoS One 10, e0142005 (2015).

    PubMed  PubMed Central  Google Scholar 

  51. Suomalainen, A. & Battersby, B. J. Mitochondrial diseases: the contribution of organelle stress responses to pathology. Nat. Rev. Mol. Cell Biol. 19, 77–92 (2018).

    CAS  PubMed  Google Scholar 

  52. Yoshii, S. R., Kishi, C., Ishihara, N. & Mizushima, N. Parkin mediates proteasome-dependent protein degradation and rupture of the outer mitochondrial membrane. J. Biol. Chem. 286, 19630–19640 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Horie, C., Suzuki, H., Sakaguchi, M. & Mihara, K. Characterization of signal that directs C-tail-anchored proteins to mammalian mitochondrial outer membrane. Mol. Biol. Cell 13, 1615–1625 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Hitz, C., Steuber-Buchberger, P., Delic, S., Wurst, W. & Kühn, R. Generation of shRNA transgenic mice. Methods Mol. Biol. 530, 101–129 (2009).

    CAS  PubMed  Google Scholar 

  55. Ortiz, O., Wurst, W. & Kühn, R. Reversible and tissue-specific activation of MAP kinase signaling by tamoxifen in Braf(V637)ER(T2) mice. Genesis 51, 448–455 (2013).

    CAS  PubMed  Google Scholar 

  56. Hitz, C., Wurst, W. & Kühn, R. Conditional brain-specific knockdown of MAPK using Cre/loxP regulated RNA interference. Nucl. Acids Res. 35, e90 (2007).

    PubMed  PubMed Central  Google Scholar 

  57. Breckwoldt, M. O. et al. Multiparametric optical analysis of mitochondrial redox signals during neuronal physiology and pathology in vivo. Nat. Med. 20, 555–560 (2014).

    CAS  PubMed  Google Scholar 

  58. Gorski, J. A. et al. Cortical excitatory neurons and glia, but not GABAergic neurons, are produced in the Emx1-expressing lineage. J. Neurosci. 22, 6309–6314 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Rossi, J. et al. Melanocortin-4 receptors expressed by cholinergic neurons regulate energy balance and glucose homeostasis. Cell Metab. 13, 195–204 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Barski, J. J., Dethleffsen, K. & Meyer, M. Cre recombinase expression in cerebellar purkinje cells. Genesis 28, 93–98 (2000).

    CAS  PubMed  Google Scholar 

  61. Fünfschilling, U. & Reichardt, L. F. Cre-mediated recombination in rhombic lip derivatives. Genesis 33, 160–169 (2002).

    PubMed  PubMed Central  Google Scholar 

  62. Gregorian, C. et al. Pten deletion in adult neural stem/progenitor cells enhances constitutive neurogenesis. J. Neurosci. 29, 1874–1886 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Gong, S. et al. Targeting Cre recombinase to specific neuron populations with bacterial artificial chromosome constructs. J. Neurosci. 27, 9817–9823 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Gong, S. et al. A gene expression atlas of the central nervous system based on bacterial artificial chromosomes. Nature 425, 917–925 (2003).

    CAS  PubMed  Google Scholar 

  65. Busche, M. A. et al. Clusters of hyperactive neurons near amyloid plaques in a mouse model of alzheimer’s disease. Science 321, 1686–1689 (2008).

    CAS  PubMed  Google Scholar 

  66. Gurney, M. E. et al. Motor neuron degeneration in mice that express a human Cu,Zn superoxide dismutase mutation. Science 264, 1772–1775 (1994).

    CAS  PubMed  Google Scholar 

  67. Rodriguez, C. I. et al. High-efficiency deleter mice show that FLPe is an alternative to Cre-loxP. Nat. Genet. 25, 139–140 (2000).

    CAS  PubMed  Google Scholar 

  68. Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Brill, M. S. et al. Branch-specific microtubule destabilization mediates axon branch loss during neuromuscular synapse elimination. Neuron 92, 845–856 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Kleele, T. et al. An assay to image neuronal microtubule dynamics in mice. Nat. Commun. 5, 4827 (2014).

    CAS  PubMed  Google Scholar 

  71. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    CAS  PubMed  Google Scholar 

  72. Wettmarshausen, J. & Perocchi, F. Isolation of functional mitochondria from cultured cells and mouse tissues. Methods Mol. Biol. 1567, 15–32 (2017).

    CAS  PubMed  Google Scholar 

  73. Franko, A. et al. Efficient isolation of pure and functional mitochondria from mouse tissues using automated tissue disruption and enrichment with anti-TOM22 magnetic beads. PLoS One 8, e82392 (2013).

    PubMed  PubMed Central  Google Scholar 

  74. Sims, N. R. & Anderson, M. F. Isolation of mitochondria from rat brain using percoll density gradient centrifugation. Nat. Protoc. 3, 1228–1239 (2008).

    CAS  PubMed  Google Scholar 

  75. Wang, X. et al. Isolation of brain mitochondria from neonatal mice. J. Neurochem. 119, 1253–1261 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Watkins, P. A., Ferrell, E. V. Jr., Pedersen, J. I. & Hoefler, G. Peroxisomal fatty acid beta-oxidation in HepG2 cells. Arch. Biochem. Biophys. 289, 329–336 (1991).

    CAS  PubMed  Google Scholar 

  77. Demarquoy, J. & Le Borgne, F. Crosstalk between mitochondria and peroxisomes. World J. Biol. Chem. 6, 301–309 (2015).

    PubMed  PubMed Central  Google Scholar 

  78. Wiśniewski, J. R., Zougman, A., Nagaraj, N. & Mann, M. Universal sample preparation method for proteome analysis. Nat. Methods 6, 359–362 (2009).

    PubMed  Google Scholar 

  79. Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).

    CAS  PubMed  Google Scholar 

  80. Cox, J. et al. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol. Cell Proteom. 13, 2513–2526 (2014).

    CAS  Google Scholar 

  81. Vizcaino, J. A. et al. update of the PRIDE database and its related tools. Nucl. Acids Res. 44, D447–456 (2016).

    CAS  PubMed  Google Scholar 

  82. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    CAS  PubMed  Google Scholar 

  83. Cox, J. & Mann, M. 1D and 2D annotation enrichment: a statistical method integrating quantitative proteomics with complementary high-throughput data. BMC Bioinform. 13 (Suppl. 16), S12 (2012).

    CAS  Google Scholar 

  84. Goldberg, T. et al. LocTree3 prediction of localization. Nucl. Acids Res. 42, W350–W355 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Wang, J., Vasaikar, S., Shi, Z., Greer, M. & Zhang, B. WebGestalt 2017: a more comprehensive, powerful, flexible and interactive gene set enrichment analysis toolkit. Nucl. Acids Res. 45, W130–W137 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Kanehisa, M., Goto, S., Sato, Y., Furumichi, M. & Tanabe, M. KEGG for integration and interpretation of large-scale molecular data sets. Nucl. Acids Res. 40, D109–D114 (2011).

    PubMed  PubMed Central  Google Scholar 

  87. Shin, J. B. et al. Molecular architecture of the chick vestibular hair bundle. Nat. Neurosci. 16, 365–374 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Uchida, Y. & Takahashi, H. Rapid detection of Abeta deposits in APP transgenic mice by Hoechst 33342. Neurosci. Lett. 448, 279–281 (2008).

    CAS  PubMed  Google Scholar 

  89. Tillberg, P. W. et al. Protein-retention expansion microscopy of cells and tissues labeled using standard fluorescent proteins and antibodies. Nat. Biotechnol. 34, 987–992 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Budak, N. Budak and S. Taskin for animal husbandry and A. Berghofer, A. Graupner, Y. Hufnagel, K. Wullimann and M. Schetterer for technical and administrative support. We thank U. Fünfschilling (Max Planck Institute Göttingen) for providing Gabra6:Cre mice, D. Crane (Griffith University) for the Pex14 antibody and N. Mizushima (Addgene plasmid #38249) for the pMXs-IP GFP–Omp25 plasmid. We are grateful to L. Jiang for help with bioinformatics analyses, Y.-H. Tai for neonatal virus injection and B. Zott for APPPS1 breeding. We thank L. Godinho, M. Schuldiner and O. Schuldiner for reading an earlier version of this article. This work was supported by the Deutsche Forschungsgemeinschaft through the Munich Center for Systems Neurology (SyNergy; grant no. EXC 1010 to A.K., T.K., S.F.L., T.M., F.P. and W.W.); the Center for Integrated Protein Science Munich (grant no. EXC 114 to A.K. and T.M.); the Collaborative Research Centers (CRC870 to T.M., CRC1054 to T.K. and CRC-TR128 to T.K.); research unit grant no. FOR2290 to S.F.L. and research grant no. Mi694/7-1/8-1 to T.M. F.P. and J.W. were supported by the Emmy-Noether program of the Deutsche Forschungsgemeinschaft (grant no. Pe2053/1-1 to F.P.). Further support came from the European Research Council under the European Union’s Seventh Framework Program (grant no. FP/2007-2013; European Research Council Grant Agreement nos. CoG 616791 to T.M. and CoG 647215 to T.K.) and the German Center for Neurodegenerative Diseases (DZNE Munich to S.F.L., T.M. and W.W.). L.T. was supported by an EMBO Long-Term Fellowship (no. EMBO ALTF 108-2013). Further support came from the Centers of Excellence in Neurodegeneration and the Helmholtz-Israel Program to S.F.L., the Swiss National Science Formation and HHS Foundation to D.M., the German Federal Ministry of Education and Research (BMBF) through ‘T-B interaction in NMO’ to T.K. and ‘Mitochondrial endophenotypes of Morbus Parkinson’ (grant no. 031A430E) to W.W. A.K. is supported by a Senior Hertie Professorship of Neuroscience. T.M. is supported by the Institute for Advanced Study, Technical University of Munich (Focus Group ‘Subcellular Dynamics in Neurons’).

Author information

Authors and Affiliations

Authors

Contributions

C.F., L.T. and T.M. devised the study. L.T., T.M., O.O., R.K. and W.W. designed and generated the MitoTag mice, which C.F. and L.T. characterized. C.F. designed the immunocapture protocol (with support from J.W. and F.P.) and performed most of the isolations. S.A.M. and S.F.L. performed sample preparation, mass spectrometry and primary data analysis. C.F. further analyzed the proteomics datasets. L.T. performed bioenergetics measurements and corresponding isolations (with support from J.W. and F.P.). C.F. performed calcium uptake assays (with support from F.P.). C.F., S.H. and T.K. performed and analyzed flow cytometry. C.F. performed western blot analysis and immunofluorescence stainings of candidates. N.S. and C.F. obtained and analyzed electron microscopy data. C.F. characterized the Rmdn3−/− and GC-specific McuFL/FL mouse models. J.H., R.M.K. and A.K. provided pilot experiments for single-cell characterization in the cerebellum. A.K. provided APPPS1 mice. I.W. and D.M. performed immunofluorescence staining on human tissue. C.F., L.T. and T.M. wrote the paper with input from all authors.

Corresponding author

Correspondence to Thomas Misgeld.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Nature Neuroscience thanks Robert Friedlander, Daniel McClatchy and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Supplementary Figs. 1–18 and Supplementary Tables 1–4.

Reporting Summary

Supplementary Data

Proteomics data from cell-type-specific mitochondria: Purkinje cells, granule cells and astrocytes.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fecher, C., Trovò, L., Müller, S.A. et al. Cell-type-specific profiling of brain mitochondria reveals functional and molecular diversity. Nat Neurosci 22, 1731–1742 (2019). https://doi.org/10.1038/s41593-019-0479-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-019-0479-z

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research