Elsevier

Neuromuscular Disorders

Volume 22, Issue 11, November 2012, Pages 1015-1024
Neuromuscular Disorders

Sustained expression and safety of human GNE in normal mice after gene transfer based on AAV8 systemic delivery

https://doi.org/10.1016/j.nmd.2012.03.013Get rights and content

Abstract

GNE myopathy is an autosomal recessive adult onset disorder caused by mutations in the GNE gene. GNE encodes the bifunctional enzyme UDP-N-acetylglucosamine 2-epimerase/N-acetyl mannosamine kinase, the key enzyme in the biosynthesis pathway of sialic acid. Additional functions for GNE have been described recently, but the mechanism leading from GNE mutation to this myopathy is unclear. Therefore a gene therapy approach could address all potential defects caused by GNE mutations in muscle. We show that AAV8 viral vectors carrying wild type human GNE cDNA are able to transduce murine muscle cells and human GNE myopathy-derived muscle cells in culture and to express the transgene in these cells. Furthermore, the intravenous administration of this viral vector to healthy mice allows expression of the GNE transgene mRNA and of the coexpressed luciferase protein, for at least 6 months in skeletal muscles, with no clinical or pathological signs of focal or general toxicity, neither from the virus particles nor from the wild type human GNE overexpression. Our results support the future use of an AAV8 based vector platform for a safe and efficient therapy of muscle in GNE myopathy.

Introduction

Mutations in the UDP-N-acetylglucosamine 2-epimerase/N-acetyl mannosamine kinase encoding gene (GNE) lead to an adult onset recessive myopathy [1]. This progressive myopathy with typical distal onset and quadriceps sparing received different names in the past: hereditary inclusion body myopathy (HIBM) [2], quadriceps sparing myopathy [3], and distal myopathy with rimmed vacuoles (DMRV, Nonaka’s disease) [4]. Upon recently it has been agreed by the vast majority of researchers and clinicians dealing with this disease to designate this condition by a single name: GNE myopathy, in order to emphasize that all three commonly used names point to the same single entity: myopathy due to GNE mutations. This disease is common particularly in the Persian Jewish community (estimated prevalence of 1:1500), but was also described in non Jewish families from the Middle East (all carrying the same founder mutation, M712T) [5]. The condition has however a worldwide distribution, with most patients being compound heterozygotes, carrying mutations either at the epimerase domain, at the kinase domain, or one in each domain of the GNE gene [6]. It is particularly frequent in Japan with two founders and many other less frequent mutations involving again both domains of the gene [7].

Although the role of GNE as a key enzyme in the biosynthetic pathway of sialic acid has been thoroughly investigated [8], the process by which the mutations in the enzyme lead to muscle disease is not understood. The only animal model relevant to GNE myopathy is a transgenic mouse model generated on a GNE-/- background and overexpressing the D176V GNE missense mutation occurring in the epimerase domain of the enzyme (a founder mutation in Japan) [9]. This mouse shows a strong hyposialylation of most organs in vivo, and recapitulates many histopathological features of GNE myopathy. Dietary supply of sialic acid or its metabolic intermediate ManNAc could partially prevent the development of this pathology, indicating that sialic acid deficiency is part of the disease pathomechanism [10]. Recently, additional functions have been ascribed to GNE in muscle cells, such as its binding properties to alpha actinin, its localization in the Z disk [11] and its role in cytoskeleton organization [12], suggesting that other pathways may be affected in the pathophysiology of GNE myopathy. If indeed other mechanisms contribute to the development of GNE myopathy, metabolic supplementation may not correct them all.

To address more comprehensively all the potential defects occurring in muscle tissue as the result of GNE mutations, we have taken a genetic approach as a mean to treat this disorder, and have used the adeno associated virus type 8 (AAV8) based system as a GNE delivery tool. Accumulating evidence points to recombinant AAV vectors being the most promising gene delivery tool for muscle diseases [13]. Furthermore a preclinical proof of efficacy of this treatment has been shown in the GNE myopathy mouse model [14]. In the present study we have evaluated the potential of the virus carrying the human wild type GNE gene to infect human GNE myopathy derived primary muscle cells, as well as its safety and extent of expression up to 6 months after intravenous administration to mice.

Section snippets

GNE cloning and virus production

To produce AAV8 viral vectors carrying the human GNE gene (AAV8-hGNE), we have generated GNE cDNA by PCR from our previously generated N-terminal 3XFLAG-CMV-10 GNE vector [11] and subsequently cloned it in the pCMV-Luciferase-eGFP vector (pZac2.1-luc-IRES-eGFP, supplied by Penn Vector Core at University of Pennsylvania) by replacing the luciferase gene at EcoRI/BamHI sites (Supplementary Fig. S1). Small scale viral vector for in vitro studies was produced by triple transfection into HEK293

Infection of C2C12 cells

We have cloned human GNE cDNA in a vector containing AAV2 packaging signals and GFP (Supplementary Fig. S1), and produced small scale AAV8/hGNE-IRES-GFP virus by triple transfection of HEK293 cells as described in the methods section. In order to evaluate the potential of AAV8/hGNE-IRES-GFP virus to infect muscle cells, C2C12 murine muscle cell line was infected with the virus (106 infectious particles/ml) and analyzed for expression. GFP was detected in about 12% of the cells after 2 days and

Discussion

In these experiments we have shown that AAV8 can transduce GNE myopathy muscle cells in culture, despite their potential hyposialylated state. This finding is particularly important if we consider AAV8 mediated therapy for a condition with presumed partial hyposialylation like GNE myopathy, since studies have established that some AAV virus types infect cells through sialylated receptors [21]. It should be mentioned that recent reports describe that AAV9 is more efficient when receptors are

Acknowledgements

We thank Michael Zeira for his help with the FACS analysis, Hanna Wald and Amnon Peled for help in the IP-10 assay, Eduardo Ayuso and Fatima Bosch for supplying virus for our pilot studies and for fruitful discussions, Evelyne Zeira for help with luciferase imaging. This research project was supported by the Neuromuscular Disease Foundation (NDF), Hadassah Southern California Haifa and Malka chapters, and in part by a grant from USAID’s American Schools and Hospitals Abroad (ASHA) Program for

References (37)

  • I. Eisenberg et al.

    The UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase gene is mutated in recessive hereditary inclusion body myopathy

    Nat Genet

    (2001)
  • V. Askanas et al.

    Newest approaches to diagnosis of sporadic inclusion body myositis and hereditary inclusion body myopathies, including molecular-pathologic similarities to Alzheimer disease

  • I. Nonaka et al.

    Familial distal myopathy with rimmed vacuole and lamellar (myeloid) body formation

    J Neurol Sci

    (1981)
  • I. Eisenberg et al.

    Mutation spectrum of the GNE gene in hereditary inclusion body myopathy sparing the quadriceps

    Hum Mutat

    (2003)
  • A. Arai et al.

    A novel mutation in the GNE gene and a linkage disequilibrium in Japanese pedigrees

    Ann Neurol

    (2002)
  • M.C. Malicdan et al.

    A Gne knockout mouse expressing human GNE D176V mutation develops features similar to distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy

    Hum Mol Genet

    (2007)
  • M.C. Malicdan et al.

    Prophylactic treatment with sialic acid metabolites precludes the development of the myopathic phenotypein the DMRV-hIBM mouse model

    Nat Med

    (2009)
  • S. Amsili et al.

    UDP-N acetyl glucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) binds to alpha-actinin 1: novel pathways in skeletal muscle?

    PLoS One

    (2008)
  • Cited by (26)

    • Role of HSP70 chaperone in protein aggregate phenomenon of GNE mutant cells: Therapeutic lead for GNE Myopathy

      2022, International Journal of Biochemistry and Cell Biology
      Citation Excerpt :

      Sialic acid extended release (SA-ER) and aceneuramic acid extended release (Ace-ER) administration in clinical trials patients failed due to lack of statistical significance in cohorts of patient data (Lochmüller et al., 2019). The AAV viral vector-based delivery of GNE is also being explored in transgenic mice for therapy (Mitrani-Rosenbaum et al., 2022, 2012). However, efforts are needed in the direction to unravel the pathomechanism of the disease and identify drug targets.

    • Congenital Disorders of Glycosylation

      2021, Comprehensive Glycoscience: Second Edition
    • Treatment and Management of Muscular Dystrophies

      2021, Neuromuscular Disorders: Treatment and Management
    • Therapeutic approaches in Congenital Disorders of Glycosylation (CDG) involving N-linked glycosylation: an update

      2020, Genetics in Medicine
      Citation Excerpt :

      AAVs are thought to be a relatively safe and efficient technology applicable to a wide range of target tissues, although AAV immune responses are observed in human.72 AAV therapy in CDG has been limited to GNE-CDG in mice73 and human primary muscle cells,74 where AAV therapy resulted in coexpression of wild-type and mutated GNE transcripts. Nonviral transgene delivery methods include zinc-finger nucleases, TALENs, and CRISPR/Cas9 technology.

    View all citing articles on Scopus
    View full text