Conditional expression of human β-hexosaminidase in the neurons of Sandhoff disease rescues mice from neurodegeneration but not neuroinflammation
8 pages
English

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris

Conditional expression of human β-hexosaminidase in the neurons of Sandhoff disease rescues mice from neurodegeneration but not neuroinflammation

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus
8 pages
English
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus

Description

This study evaluated whether GM 2 ganglioside storage is necessary for neurodegeneration and neuroinflammation by performing β-hexosaminidase rescue experiments in neurons of HexB −/− mice. We developed a novel mouse model, whereby the expression of the human HEXB gene was targeted to neurons of HexB −/− mice by the Thy1 promoter. Despite β-hexosaminidase restoration in neurons was sufficient in rescuing HexB −/− mice from GM 2 neuronal storage and neurodegeneration, brain inflammation persisted, including the presence of large numbers of reactive microglia/macrophages due to persisting GM 2 presence in this cell type. In conclusion, our results suggest that neuroinflammation is not sufficient to elicit neurodegeneration as long as neuronal function is restored.

Sujets

Informations

Publié par
Publié le 01 janvier 2012
Nombre de lectures 10
Langue English
Poids de l'ouvrage 1 Mo

Extrait

Kyrkanideset al. Journal of Neuroinflammation2012,9:186 http://www.jneuroinflammation.com/content/9/1/186
R E S E A R C H
JOURNAL OF NEUROINFLAMMATION
Open Access
Conditional expression of human βhexosaminidase in the neurons of Sandhoff disease rescues mice from neurodegeneration but not neuroinflammation 1,2* 2 3 3 4 Stephanos Kyrkanides , Sabine M Brouxhon , Ross H Tallents , Jennie H Miller , John A Olschowka 4 and M Kerry OBanion
Abstract This study evaluated whether GM2ganglioside storage is necessary for neurodegeneration and neuroinflammation /by performingβhexosaminidase rescue experiments in neurons of HexB mice. We developed a novel mouse /model, whereby the expression of the human HEXB gene was targeted to neurons of HexB mice by the Thy1 /promoter. Despiteβmice from GMhexosaminidase restoration in neurons was sufficient in rescuing HexB 2 neuronal storage and neurodegeneration, brain inflammation persisted, including the presence of large numbers of reactive microglia/macrophages due to persisting GM2presence in this cell type. In conclusion, our results suggest that neuroinflammation is not sufficient to elicit neurodegeneration as long as neuronal function is restored. Keywords:GM2gangliosidosis,βhexosaminidase, Mouse, Neuron, Sandhoff disease, Transgenic
Introduction The pathognomonic feature of Sandhoff disease is GM2 ganglioside storage primarily in neurons. Catabolism of GM2ganglioside in mammalian cells is undertaken by βhexosaminidase, a lysosomal acidic hydrolase. Struc turally,βhexosaminidase (HEX) is comprised of two subunits,αandβ, and exists in three isoforms HEXA (α/βheterodimer), HEXB (β/βhomodimer) and HEXS (α/αhomodimer). In humans, HEXA (α/β) catabolizes GM2presented by a third protein named GM2activator. Each subunit,αandβ, is encoded by a distinct gene, HEXAandHEXB, located on human chromosomes 15 and 5, respectively. Human patients withHEXA(TaySachs) orHEXB(Sandhoff ) mutations develop storage of GM2 ganglioside in the lysosomes due to the lack of HEXA (α/β) enzyme activity [1]. Although HEXA enzyme is pre sent in all cell types and tissues, neurons are characterized
* Correspondence: kyrkanides@gmail.com 1 Department of Childrens Dentistry, Stony Brook University, Stony Brook, NY 118948701, USA 2 Health Science Center, Stony Brook University, Stony Brook, NY 117948701, USA Full list of author information is available at the end of the article
by remarkably higher concentrations of gangliosides than other cell types and therefore are highly susceptible to GM2lysosomal storage. Similarly to humans, two murine genes encoding forβhexosaminidase have been identified: HexAandHexB[1]. Targeted deletion of theHexAlocus resulted in the development of a mouse phenotype that showed only a mild degree of the expected pathology and lack of any neurological findings [1]. In contrast, disrup tion of the murineHexBlocus resulted in a mouse pheno type that closely resembled that of the human disease. The mice displayed storage of GM2ganglioside in the cen tral nervous system (CNS), and neurons with membran ous cytoplasmic bodies similar to those in TaySachs and Sandhoff patients [1,2]. The phenotypic variation between humans and mice appears to result from differences in the ganglioside degradation pathway between the species. It has been proposed that a second ganglioside degrad ation pathway exists in the mouse, whereby GM2can be, at least in the absence of HEXA (α/β), metabolized by a murine sialidase to asialoGM2 and subsequently catabo lized by HEXB (β/β). However, human sialidases cannot metabolize GM2ganglioside. Therefore, in the mouse HexB disruption results in GM2gangliosidosis, whereas in
© 2012 Kyrkanides et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
  • Univers Univers
  • Ebooks Ebooks
  • Livres audio Livres audio
  • Presse Presse
  • Podcasts Podcasts
  • BD BD
  • Documents Documents