Cookies on this website

We use cookies to ensure that we give you the best experience on our website. If you click 'Accept all cookies' we'll assume that you are happy to receive all cookies and you won't see this message again. If you click 'Reject all non-essential cookies' only necessary cookies providing core functionality such as security, network management, and accessibility will be enabled. Click 'Find out more' for information on how to change your cookie settings.

Activation of peroxisome proliferator-activated receptor-gamma (PPARgamma) signaling after stroke may reduce brain injury, but this effect will depend on the levels of receptor and cofactors. Here, we showed that the direct effect of PPARgamma signaling to protect neurons from ischemic injury requires a novel cofactor LMO4, because this effect was lost in LMO4-null cortical neurons. PPARgamma agonist also failed to reduce cerebral infarction after transient focal ischemia in CaMKIIalphaCre/LMO4loxP mice with LMO4 ablated in neurons of the forebrain. Expressing LMO4 in LMO4-null cortical neurons rescued the PPARgamma-protective effect. PPARgamma signaling activates the promoter of the antioxidant gene SOD2 and this process requires LMO4. Addition of a superoxide dismutase mimetic MnTBAP [manganese(III)tetrakis(4-benzoic acid)porphyrin] bypassed the deficiency in PPARgamma signaling and was able to directly rescue LMO4-null cortical neurons from ischemic injury. Like LMO4, PPARgamma and PGC1alpha (PPARgamma coactivator 1alpha) levels in neurons are elevated by hypoxic stress, and absence of LMO4 impairs their upregulation. Coimmunoprecipitation and mammalian two-hybrid assays revealed that LMO4 interacts in a ligand-dependent manner with PPARgamma. LMO4 augments PPARgamma-dependent gene activation, in part, by promoting RXRalpha (retinoid X receptor-alpha) binding to PPARgamma and by increasing PPARgamma binding to its target DNA sequence. Together, our results identify LMO4 as an essential hypoxia-inducible cofactor required for PPARgamma signaling in neurons. Thus, upregulation of LMO4 expression after stroke is likely to be an important determinant of neuron survival.

Original publication

DOI

10.1523/JNEUROSCI.2897-08.2008

Type

Journal article

Journal

J Neurosci

Publication Date

19/11/2008

Volume

28

Pages

12433 - 12444

Keywords

Adaptor Proteins, Signal Transducing, Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2, Cell Death, Cell Hypoxia, Cells, Cultured, Cerebral Cortex, Disease Models, Animal, Electrophoretic Mobility Shift Assay, Embryo, Mammalian, Gene Expression Regulation, Glucose, Homeodomain Proteins, Hypoglycemic Agents, Immunoprecipitation, Infarction, Middle Cerebral Artery, Kainic Acid, LIM Domain Proteins, Mice, Mice, Knockout, N-Methylaspartate, Neurons, Oxygen, PPAR gamma, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Rosiglitazone, Signal Transduction, Superoxide Dismutase, Thiazolidinediones, Trans-Activators, Transcription Factors