13.01.2014 Views

0-TESTO COMPLETO.pdf - Fondazione Santa Lucia

0-TESTO COMPLETO.pdf - Fondazione Santa Lucia

0-TESTO COMPLETO.pdf - Fondazione Santa Lucia

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

PFIZER.2 – Effects of the phosphodiesterase type X (PDE10) inhibitor on R6/2 mouse model of HD<br />

The results of our studies with rolipram provide strong theoretical support<br />

for the strategy of targeting CREB signaling to promote striatal and cortical<br />

neuron survival in HD. However, these studies do not provide a direct path<br />

to a therapy since there are no PDE4 inhibitors currently available for clinical<br />

use, as these agents suffer from severe side effect liabilities. Thus, we have<br />

turned to the investigation of another phosphodiesterase, PDE10A, which<br />

may be a particularly promising target as a potential treatment for HD [Chappie<br />

et al. 2009; Giampà et al. 2009]. PDE10A is highly expressed in the striatal<br />

medium spiny neurons [Seeger et al. 2003; Coskran et al. 2006; Xie et al.<br />

2006] vulnerable in HD, where it regulates both cAMP and cGMP signaling<br />

cascades [Siuciak et al. 2006a; Siuciak et al. 2006b]. Notably, inhibition of<br />

PDE10A with the highly specific inhibitor TP-10 results in a significant<br />

increased in CREB phosphorylation in striatum [Schmidt et al. 2008]. A<br />

recent study published by our group showed that TP-10 treatment in our rat<br />

QA model resulted in a significant sparing of striatal neurons, on one hand,<br />

and a parallel increase in activated CREB, on the other [Giampà et al. 2009].<br />

Significantly, TP-10 treatment also produced a significant sparing of cortical<br />

neurons in this model, either as a result of a primary neuroprotective effect on<br />

striatal neurons or, possibly, by a more direct effect (see further discussion<br />

below). These encouraging results lead us to continue our research by testing<br />

whether the same neuroprotective effects could be achieved by TP-10 in the<br />

well-established transgenic mouse model of HD, namely, Bates’ R6/2 mice<br />

[Mangiarini et al. 1996].<br />

EFFECT OF TP-10 TREATMENT ON THE R6/2 MICE:<br />

RESULTS TO DATE<br />

R6/2 mice and wild type littermate controls were treated daily with vehicle<br />

or TP-10 beginning at 4 weeks of age through study termination. In the<br />

R6/2 animals, TP-10 treatment increased survival time by approximately 10%.<br />

However, the more significant finding is that TP-10 treatment almost completely<br />

ameliorated the development of neurological deficits in the R6/2 mice<br />

prior to death. Specifically, at 12 weeks of age, when R6/2 mice treated with<br />

vehicle were fully symptomatic, R6/2 mice treated with TP-10 displayed<br />

almost no hind paw clasping during tail suspension, no deficit in rotarod performance,<br />

and no decrease in locomotor activity in an open field. This effect<br />

of TP-10 treatment on neurological function was reflected in a significant<br />

amelioration in brain pathology in the R6/2 mice. TP-10 treatment largely prevented<br />

the gross decrease in brain volume, and the dramatic reduction in striatal<br />

cell number and cell size that was characteristically observed in the vehicle<br />

treated R6/2 animals. TP-10 treatment also significantly reduced the<br />

degree of microglial activation that occurs in response to the mutant huntingtin-induced<br />

brain damage.<br />

In summary, results to date indicate that TP-10 treatment has a dramatic<br />

therapeutic effect to prevent the loss of function caused by the Huntington’s<br />

mutation in the R6/2 mice. In this grant renewal, we continue this investigation<br />

with four Specific Aims. These are:<br />

2009 667

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!