Similarly, P-Akt colocalized with phospho-Histone H3, a marker of

Similarly, P-Akt colocalized with phospho-Histone H3, a marker of M phase, in dividing apical progenitors as well as GLAST, a marker of radial glial cells (data not shown). Because these apical progenitor cells give rise to both neurons and glia, this localization is consistent with activation of AKT3 in both neurons and glia. Abnormal AKT function would be consistent with the MRI patterns and neuropathological studies ( Figures 1 and 2), which show abnormal organization MK-1775 supplier of neurons in the cortex and abnormal MRI signal characteristics of white matter. Our data suggest that activation of AKT3, either by duplication or by point mutation, contributes to hemispheric

brain overgrowth. Two of our cases (the point mutation and one partial trisomy) are confirmed to be de novo, somatic mutations, undetectable in blood, and although nonbrain tissues were not available from the other partial trisomy case, this is likely

to be a somatic mutation as well, because http://www.selleckchem.com/products/Imatinib-Mesylate.html individuals reported with constitutional trisomy 1q, even a portion of 1q, show dysmorphic features and, in nearly all cases, early lethality ( Mark et al., 2005, Mefford et al., 2008 and Patel et al., 2009). We postulate that increasing AKT3 dosage and activation of AKT3 would have the same effect in the setting of a somatic mutation. Interestingly, HMG has not been reported in the constitutional trisomy cases, even those that have partial trisomy including AKT3. It is possible that HMG might not be present in the cases with early lethality; perhaps more important, because all of the constitutional trisomy 1q cases were de novo, the trisomy may not be present in all tissues. Though we have not sampled other tissues, there was no clinical evidence of extracerebral involvement phenotypically in any of the three cases, suggesting that either the mutation was limited to the brain or activation of AKT3 in other tissues does not have phenotypic consequences. Increased rates of brain cancer are not reported new in the setting

of isolated HMG. In the cases we report here, which have not shown any form of cancer, it is likely that activation of AKT3 disrupts normal cortical development but does not result in continued dysregulated growth outside the setting of cortical progenitor cells. Further support for the role of AKT3 in controlling brain size comes from animal studies. A mouse Akt3 knockout model shows selective reduction in brain size due to decreased neuronal number and size ( Easton et al., 2005), whereas mice with an activating mutation in the kinase domain of Akt3 show larger hippocampal size and abnormal Ki67-positive ectopic neurons in the hippocampus ( Tokuda et al., 2011). Additionally, in zebrafish, overexpression of wild-type akt3 produces increased embryonic brain thickness ( Chen et al., 2011). All of these results strongly suggest that AKT3 activity dynamically regulates brain size and that increased dosage of AKT3 might increase brain size in humans.

Comments are closed.