Previous Page  21 / 34 Next Page
Information
Show Menu
Previous Page 21 / 34 Next Page
Page Background

Page 61

Notes:

conferenceseries

.com

Volume 8, Issue 2 (Suppl)

J Neurol Neurophysiol

ISSN: 2155-9562 JNN, an open access journal

Neurology 2017

March 27-29, 2017

March 27-29, 2017 Madrid, Spain

11

th

World Congress on

Neurology and Therapeutics

Plastin3 as a therapeutic target in spinal muscular atrophy

Aziza Al-Rafiah

King Abdul Aziza University, Saudi Arabia

S

pinal muscular atrophy (SMA) is a devastating childhood motor neuron disease caused by mutations in the survival motor neuron

1 gene

(SMN1). SMN1

and

SMN2

are nearly identical genes producing the survival of motor neuron (SMN) protein. SMN protein

plays a crucial role in mRNA splicing and β-actin mRNA transport along the axons. In SMA, the mutation leads to the loss of

SMN1

,

which cannot be fully compensated by the

SMN2

gene, which predominantly produces a truncated protein. The loss or reduction of

SMN protein leads to motor axonal defects and motor neuron cell death. There are currently no treatments available but therapies

have focused on increasing SMN through replacing SMN1 or increasing full length SMN from

SMN2

. The actin-binding protein

Plastin 3 (

PLS3

) has been reported as a modifier for SMA, making it a potential therapeutic target. Recently, it was shown that the

overexpression of the PLS3 gene improved axonal outgrowth in SMN- deficient motor neurons of SMA Zebra fish and cultured

motor neurons from mouse embryos. Gene therapy using viral vectors was carried out

in vitro

and

in vivo

to assess whether the

overexpression of

PLS3

could rescue neuronal loss in SMA and be developed as a therapy. The SMN☐7 mouse model produces low

levels of SMN, modelling severe SMA disease with an average lifespan of 12 days and loss of motor neurons. This study has established

that the SMNΔ7 mice have little or no detectable

PLS3

from birth, making it a good model for developing PLS3 gene therapy.

Lentiviral vectors were able to upregulate PLS3 expression in different cell lines. Transduction of NSC34 cells with LV-PLS3 vector led

to a five-fold increase in expression of PLS3 compared to controls. In smn-deficient MNs, expression of

PLS3

restored axonal length

and showed a strong neuroprotective effect. Pre-clinical in vivo proof-of-concept studies using adeno-associated virus serotype 9

(AAV9) encoding PLS3 in SMNΔ7 mice showed high transduction efficiency and overexpression of

PLS3

specifically targeted to

neurons in the central nervous system (CNS). This led to a small but significant increase of lifespan by 54%. However,

PLS3

was not

able to prevent disease onset. Although there was no improvement of phenotype, this study has demonstrated the potential use of

PLS3

as a target for gene therapy, possibly in conjunction with other modulators of disease.

aalrafiah@kau.edu.sa

Aziza Al-Rafiah, J Neurol Neurophysiol 2017, 8:2 (Suppl)

http://dx.doi.org/10.4172/2155-9562.C1.046