Neuronal Hypoxia Induces Hsp40-Mediated Nuclear Import of Type 3 Deiodinase As an Adaptive Mechanism to Reduce Cellular Metabolism

Authors:
Jo S, Kalló I, Bardóczi Z, Arrojo e Drigo R, Zeöld A, Liposits Z, Oliva A, Lemmon VP, Bixby JL, Gereben B, Bianco AC.
In:
Source: J Neurosci
Publication Date: (2012)
Issue: 32(25): 8491-500
Research Area:
Neurobiology
Cells used in publication:
SK-N-AS
Species: human
Tissue Origin: brain
Platform:
4D-Nucleofector® X-Unit
Experiment

Nucleofected SK-N-AS cell line using 4D X-unit and SF solution. Used program DN-100. Experiment done in 20uL volume using 15,000 cells per well. Summary (by Lonza): The authors present a study of induced hypoxia in rat hippocampal neurons. The main idea is that hypoxia in neurons leads to the nuclear import of type 3 deiodinase (D3). To understand the mechnanism of nuclear import and involved proteins, one step was to study the effect of overexpression or down-regulation of Hsp40. Therefore, the human neuroblastoma cell line SK-N-AS was transfected with Hsp40-expressing plasmid or Hsp40 shRNA vector using Lonza’s 4D-Nucleofector™ System with solution SF and program DN-100. Overall the authors found that the HSp40-mediated import of D3 into the nucleus of the cells is an adaptive mechanism which decreases thyroid hormone signaling and thus minimizes cell metabolism and damage caused by hypoxia.

Abstract

In neurons, the type 3 deiodinase (D3) inactivates thyroid hormone and reduces oxygen consumption, thus creating a state of cell-specific hypothyroidism. Here we show that hypoxia leads to nuclear import of D3 in neurons, without which thyroid hormone signaling and metabolism cannot be reduced. After unilateral hypoxia in the rat brain, D3 protein level is increased predominantly in the nucleus of the neurons in the pyramidal and granular ipsilateral layers, as well as in the hilus of the dentate gyrus of the hippocampal formation. In hippocampal neurons in culture as well as in a human neuroblastoma cell line (SK-N-AS), a 24 h hypoxia period redirects active D3 from the endoplasmic reticulum to the nucleus via the cochaperone Hsp40 pathway. Preventing nuclear D3 import by Hsp40 knockdown resulted an almost doubling in the thyroid hormone-dependent glycolytic rate and quadrupling the transcription of thyroid hormone target gene ENPP2. In contrast, Hsp40 overexpression increased nuclear import of D3 and minimized thyroid hormone effects in cell metabolism. In conclusion, ischemia/hypoxia induces an Hsp40-mediated translocation of D3 to the nucleus, facilitating thyroid hormone inactivation proximal to the thyroid hormone receptors. This adaptation decreases thyroid hormone signaling and may function to reduce ischemia-induced hypoxic brain damage.