Slowed progression in models of Huntington disease by adipose stem cell transplantation

ST Lee, K Chu, KH Jung, WS Im, JE Park… - Annals of Neurology …, 2009 - Wiley Online Library
ST Lee, K Chu, KH Jung, WS Im, JE Park, HC Lim, CH Won, SH Shin, SK Lee, M Kim
Annals of Neurology: Official Journal of the American Neurological …, 2009Wiley Online Library
Objective Adipose‐derived stem cells (ASCs) are readily accessible and secrete multiple
growth factors. Here, we show that ASC transplantation rescues the striatal pathology of
Huntington disease (HD) models. Methods ASCs were isolated from human subcutaneous
adipose tissue. In a quinolinic acid (QA)‐induced rat model of striatal degeneration, human
ASCs (1 million cells) were transplanted into the ipsilateral striatal border immediately after
the QA injection. In 60‐day‐old R6/2 mice transgenic for HD, ASCs (0.5 million cells) were …
Objective
Adipose‐derived stem cells (ASCs) are readily accessible and secrete multiple growth factors. Here, we show that ASC transplantation rescues the striatal pathology of Huntington disease (HD) models.
Methods
ASCs were isolated from human subcutaneous adipose tissue. In a quinolinic acid (QA)‐induced rat model of striatal degeneration, human ASCs (1 million cells) were transplanted into the ipsilateral striatal border immediately after the QA injection. In 60‐day‐old R6/2 mice transgenic for HD, ASCs (0.5 million cells) were transplanted into each bilateral striata. In in vitro experiments, we treated mutant huntingtin gene‐transfected cerebral neurons with ASC‐conditioned media.
Results
In the QA model, human ASCs reduced apomorphine‐induced rotation behavior, lesion volume, and striatal apoptosis. In R6/2 transgenic mice, transplantation of ASCs improved Rota‐Rod performance and limb clasping, increased survival, attenuated the loss of striatal neurons, and reduced the huntingtin aggregates. ASC‐transplanted R6/2 mice expressed elevated levels of peroxisome proliferator‐activated receptor γ coactivator‐1α (PGC‐1α) and reactive oxygen defense enzymes and showed activation of the Akt/cAMP‐response element‐binding proteins. ASC‐conditioned media decreased the level of N‐terminal fragments of mutant huntingtin and associated apoptosis, and increased PGC‐1α expression.
Interpretation
Collectively, ASC transplantation slowed striatal degeneration and behavioral deterioration of HD models, possibly via secreted factors. Ann Neurol 2009;66:671–681
Wiley Online Library