Transgenic neuronal overexpression reveals that stringently regulated p23 expression is critical for coordinated movement in mice

Mol Neurodegener. 2011 Dec 28:6:87. doi: 10.1186/1750-1326-6-87.

Abstract

Background: p23 belongs to the highly conserved p24 family of type I transmembrane proteins, which participate in the bidirectional protein transport between the endoplasmic reticulum and Golgi apparatus. Mammalian p23 has been shown to interact with γ-secretase complex, and modulate secretory trafficking as well as intramembranous processing of amyloid precursor protein in cultured cells. Negative modulation of β-amyloid production by p23 in cultured cell lines suggested that elevation of p23 expression in neurons might mitigate cerebral amyloid burden.

Results: We generated several lines of transgenic mice expressing human p23 in neurons under the control of Thy-1.2 promoter. We found that even a 50% increase in p23 levels in the central nervous system of mice causes post-natal growth retardation, severe neurological problems characterized by tremors, seizure, ataxia, and uncoordinated movements, and premature death. The severity of the phenotype closely correlated with the level of p23 overexpression in multiple transgenic lines. While the number and general morphology of neurons in Hup23 mice appeared to be normal throughout the brain, abnormal non-Golgi p23 localization was observed in a subset of neurons with high transgene expression in brainstem. Moreover, detailed immunofluorescence analysis revealed marked proliferation of astrocytes, activation of microglia, and thinning of myelinated bundles in brainstem of Hup23 mice.

Conclusions: These results demonstrate that proper level of p23 expression is critical for neuronal function, and perturbing p23 function by overexpression initiates a cascade of cellular reactions in brainstem that leads to severe motor deficits and other neurological problems, which culminate in premature death. The neurological phenotype observed in Hup23 mice highlights significant adverse effects associated with manipulating neuronal expression of p23, a previously described negative modulator of γ-secretase activity and β-amyloid production. Moreover, our report has broader relevance to molecular mechanisms in several neurodegenerative diseases as it highlights the inherent vulnerability of the early secretory pathway mechanisms that ensure proteostasis in neurons.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Amyloid beta-Protein Precursor / metabolism
  • Animals
  • Brain / anatomy & histology
  • Brain / metabolism
  • Cells, Cultured
  • Humans
  • Membrane Proteins / genetics
  • Membrane Proteins / metabolism*
  • Mice
  • Mice, Inbred C57BL
  • Mice, Transgenic
  • Motor Activity / physiology*
  • Movement Disorders / pathology
  • Movement Disorders / physiopathology*
  • Myelin Basic Protein / metabolism
  • Neurons / cytology
  • Neurons / metabolism*
  • Neurons / pathology
  • Nucleocytoplasmic Transport Proteins

Substances

  • Amyloid beta-Protein Precursor
  • Membrane Proteins
  • Myelin Basic Protein
  • Nucleocytoplasmic Transport Proteins
  • TMED10 protein, human