Loss of prohibitin membrane scaffolds impairs mitochondrial architecture and leads to tau hyperphosphorylation and neurodegeneration

PLoS Genet. 2012;8(11):e1003021. doi: 10.1371/journal.pgen.1003021. Epub 2012 Nov 8.

Abstract

Fusion and fission of mitochondria maintain the functional integrity of mitochondria and protect against neurodegeneration, but how mitochondrial dysfunctions trigger neuronal loss remains ill-defined. Prohibitins form large ring complexes in the inner membrane that are composed of PHB1 and PHB2 subunits and are thought to function as membrane scaffolds. In Caenorhabditis elegans, prohibitin genes affect aging by moderating fat metabolism and energy production. Knockdown experiments in mammalian cells link the function of prohibitins to membrane fusion, as they were found to stabilize the dynamin-like GTPase OPA1 (optic atrophy 1), which mediates mitochondrial inner membrane fusion and cristae morphogenesis. Mutations in OPA1 are associated with dominant optic atrophy characterized by the progressive loss of retinal ganglion cells, highlighting the importance of OPA1 function in neurons. Here, we show that neuron-specific inactivation of Phb2 in the mouse forebrain causes extensive neurodegeneration associated with behavioral impairments and cognitive deficiencies. We observe early onset tau hyperphosphorylation and filament formation in the hippocampus, demonstrating a direct link between mitochondrial defects and tau pathology. Loss of PHB2 impairs the stability of OPA1, affects mitochondrial ultrastructure, and induces the perinuclear clustering of mitochondria in hippocampal neurons. A destabilization of the mitochondrial genome and respiratory deficiencies manifest in aged neurons only, while the appearance of mitochondrial morphology defects correlates with tau hyperphosphorylation in the absence of PHB2. These results establish an essential role of prohibitin complexes for neuronal survival in vivo and demonstrate that OPA1 stability, mitochondrial fusion, and the maintenance of the mitochondrial genome in neurons depend on these scaffolding proteins. Moreover, our findings establish prohibitin-deficient mice as a novel genetic model for tau pathologies caused by a dysfunction of mitochondria and raise the possibility that tau pathologies are associated with other neurodegenerative disorders caused by deficiencies in mitochondrial dynamics.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis
  • Caenorhabditis elegans Proteins
  • Genome, Mitochondrial
  • Membrane Fusion
  • Mice
  • Mitochondria* / genetics
  • Mitochondria* / metabolism
  • Mitochondria* / pathology
  • Mitochondrial Membranes / metabolism
  • Morphogenesis
  • Neurodegenerative Diseases* / genetics
  • Neurodegenerative Diseases* / metabolism
  • Neurodegenerative Diseases* / pathology
  • Neurons* / metabolism
  • Neurons* / pathology
  • Optic Atrophy, Autosomal Dominant* / genetics
  • Optic Atrophy, Autosomal Dominant* / metabolism
  • Phosphorylation
  • Prohibitins
  • Repressor Proteins* / genetics
  • Repressor Proteins* / metabolism
  • tau Proteins / genetics
  • tau Proteins / metabolism

Substances

  • Caenorhabditis elegans Proteins
  • Phb2 protein, mouse
  • Prohibitins
  • Repressor Proteins
  • prohibitin 2, C elegans
  • tau Proteins

Grants and funding

This work was supported by grants of the Deutsche Forschungsgemeinschaft to TL (SFB635, C4) and EIR (RU1653/1-1) and the European Research Council (AdG No. 233078) to TL. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.