Huntington‘s Disease (HD) is an inherited neurodegenerative disorder characterized by dyskinesia, cognitive impairment and emotional disturbances and associated with a progressive neurodegeneration of medium-spiny neurons in the striatum and with the presence of intracellular Huntingtin (Htt) aggregates in various area of the brain. The mutation responsible for HD has been identified as a CAG expansion in the Htt gene, which is translated into a polyQ stretch at the protein level. Recombinant Adeno Associated Viral (AAV) vectors have been used successfully to transfer genes in a variety of tissues, including the brain, in adult animals. In the present study we have used rAAV9, charged with Exon 1 Htt carrying 17 and 138 CAG repeats (wild type- or m-Htt, respectively) to set up and optimize both in vitro and in vivo model of Huntington‘s Disease. For in vitro model, AAV9-Ex1-AcGFP-Q138 or, as control, AAV9-Ex1-AcGFP-Q17 viral preparations have been added to pure cortical neurons prepared from E18 rat embryos and plated in 96-well plates in conditions optimized for imaging analysis with BD Pathway 435. The infection led to a progressive formation of EM-48 positive aggregates, and subsequent neuronal suffering, measured as a decrease of NeuN- and phosphoCREB-positive cells (about 50% decrease at 4 weeks post-infection). For in vivo model, AAV9-Ex1-AcGFP-Q138 viral preparations have been injected in the right striatum of female rats. The controlateral striatum was injected with AAV9-Ex1-AcGFP-Q17 and used as control. Twenty-one days after surgery animals were processed for immunohistochemical analysis on brain slices. The Q138 injection induced the formation of AcGFP (and EM48) positive Htt aggregates in the entire striatal area, increased GFAP and microglial activation with respect to Q17 injected striatum, and neurodegeneration, as evidenced by a decrease in ChAT- and NeuN-positive neurons in the Q138-injected striatum with respect to Q17. The efficacy of sodium butyrate and Cystamine were tested in this model. The AAV9-Ex1-AcGFP-Q138 HD models may represent a useful preclinical model for the evaluation of neuroprotective approaches in HD.

Recombinant Adeno Associated Viral (AAV) vector delivery of mutant huntingtin as rat model for in vitro and in vivo studies.

MIRAGLIOTTA, VINCENZO;
2011-01-01

Abstract

Huntington‘s Disease (HD) is an inherited neurodegenerative disorder characterized by dyskinesia, cognitive impairment and emotional disturbances and associated with a progressive neurodegeneration of medium-spiny neurons in the striatum and with the presence of intracellular Huntingtin (Htt) aggregates in various area of the brain. The mutation responsible for HD has been identified as a CAG expansion in the Htt gene, which is translated into a polyQ stretch at the protein level. Recombinant Adeno Associated Viral (AAV) vectors have been used successfully to transfer genes in a variety of tissues, including the brain, in adult animals. In the present study we have used rAAV9, charged with Exon 1 Htt carrying 17 and 138 CAG repeats (wild type- or m-Htt, respectively) to set up and optimize both in vitro and in vivo model of Huntington‘s Disease. For in vitro model, AAV9-Ex1-AcGFP-Q138 or, as control, AAV9-Ex1-AcGFP-Q17 viral preparations have been added to pure cortical neurons prepared from E18 rat embryos and plated in 96-well plates in conditions optimized for imaging analysis with BD Pathway 435. The infection led to a progressive formation of EM-48 positive aggregates, and subsequent neuronal suffering, measured as a decrease of NeuN- and phosphoCREB-positive cells (about 50% decrease at 4 weeks post-infection). For in vivo model, AAV9-Ex1-AcGFP-Q138 viral preparations have been injected in the right striatum of female rats. The controlateral striatum was injected with AAV9-Ex1-AcGFP-Q17 and used as control. Twenty-one days after surgery animals were processed for immunohistochemical analysis on brain slices. The Q138 injection induced the formation of AcGFP (and EM48) positive Htt aggregates in the entire striatal area, increased GFAP and microglial activation with respect to Q17 injected striatum, and neurodegeneration, as evidenced by a decrease in ChAT- and NeuN-positive neurons in the Q138-injected striatum with respect to Q17. The efficacy of sodium butyrate and Cystamine were tested in this model. The AAV9-Ex1-AcGFP-Q138 HD models may represent a useful preclinical model for the evaluation of neuroprotective approaches in HD.
File in questo prodotto:
Non ci sono file associati a questo prodotto.

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11568/150934
 Attenzione

Attenzione! I dati visualizzati non sono stati sottoposti a validazione da parte dell'ateneo

Citazioni
  • ???jsp.display-item.citation.pmc??? ND
  • Scopus ND
  • ???jsp.display-item.citation.isi??? ND
social impact