Direct central nervous system delivery provides enhanced protection following vector mediated gene replacement in a severe model of Spinal Muscular Atrophy

https://doi.org/10.1016/j.bbrc.2011.11.121Get rights and content

Abstract

Spinal Muscular Atrophy (SMA), an autosomal recessive neuromuscular disorder, is the leading genetic cause of infant mortality. SMA is caused by the homozygous loss of Survival Motor Neuron-1 (SMN1). SMA, however, is not due to complete absence of SMN, rather a low level of functional full-length SMN is produced by a nearly identical copy gene called SMN2. Despite SMN’s ubiquitous expression, motor neurons are preferentially affected by low SMN levels. Recently gene replacement strategies have shown tremendous promise in animal models of SMA. In this study, we used self-complementary Adeno Associated Virus (scAAV) expressing full-length SMN cDNA to compare two different routes of viral delivery in a severe SMA mouse model. This was accomplished by injecting scAAV9-SMN vector intravenously (IV) or intracerebroventricularly (ICV) into SMA mice. Both routes of delivery resulted in a significant increase in lifespan and weight compared to untreated mice with a subpopulation of mice surviving more than 200 days. However, the ICV injected mice gained significantly more weight than their IV treated counterparts. Likewise, survival analysis showed that ICV treated mice displayed fewer early deaths than IV treated animals. Collectively, this report demonstrates that route of delivery is a crucial component of gene therapy treatment for SMA.

Highlights

► Low titer rescue of SMA mice. ► ICV delivery provides greater phenotypic rescue in SMA with low titer scAAV9-SMN injection. ► Extension in survival greater than 100 days for scAAV9-SMN injected mice.

Introduction

Spinal Muscular Atrophy (SMA) is caused by the homozygous loss of Survival Motor Neuron-1, SMN1 [1], [2]. The human genome contains two nearly identical SMN genes, SMN1 and SMN2, however, only SMN1 functions as the disease-determining gene [3], [4]. SMN1 and SMN2 differ by a silent C to T transition at the 5′ end of exon 7 [5], [6]. This difference alters the alternative pre-mRNA splicing ratios from the two genes, resulting in high levels of full-length product from SMN1, whereas SMN2 produces low levels of full-length SMN and an abundant alternatively spliced isoform, SMNΔ7. The truncated isoform is unstable and cannot compensate for the loss of SMN1 [3]. Despite the ubiquitous expression of SMN, preferential loss of motor neurons occurs in SMA. Because SMA is monogenic, vector-based gene replacement of SMN1 is an attractive option for the treatment of SMA. Encouraging reports have been published using a relatively severe model of SMA called SMNΔ7. These mice lack endogenous mouse Smn, but express the human SMN2 gene and the cDNA encoding the alternatively spliced isoform produced by SMN2, SMNΔ7 (Smn−/−; SMN2+/+; SMNΔ7+/+) [7]. Untreated SMNΔ7 animals live approximately 14 days with disease symptoms becoming overtly apparent around day 7 [7]. Delivery of full-length SMN cDNA to SMNΔ7 neonates using scAAV8 or scAAV9 vectors resulted in significant extensions in survival ranging from an average of 60–200+ days [8], [9], [10], [11], with some treated mice displaying a full rescue in terms of lifespan and motor function. However, it remains unclear whether the different injection paradigms or the vector serotype was the primary cause for the differences in the degree of phenotypic rescue.

In this report we utilized a scAAV9-SMN vector and examined two routes of injection in neonatal SMNΔ7 mice [12]. Pups received injections of 2 × 1010 viral genomes via the facial vein (IV) or directly into the brain ventricles (ICV) on postnatal day 2 (PND2). We demonstrate that at this relatively low viral titer, animals receiving ICV injections gained significantly more weight and lived longer than animals receiving IV injections. As expected, animals receiving ICV injections also had higher SMN protein levels in the brain and lumbar spinal cord as compared to IV injected animals. From these results, we conclude that the route of injection for scAAV9-SMN has a significant impact upon the degree of phenotypic rescue and sheds light upon the development of disease and potential therapeutic implications.

Section snippets

Genotyping and mouse handling

Animals were handled according to the University of Missouri Animal Care and Use Committee approved Protocols. Mice heterozygous for mSmn (Smn+/−; SMN2+/+; SMNΔ7+/+) were interbred to generate experimental SMA cohorts (Smn−/−; SMN2+/+; SMNΔ7+/+). The day of birth was counted as PND1 and the neonates were genotyped within 24 h. Animals were genotyped using PCR conditions as previously described [13]. SMA mice were raised with two heterozygous siblings. Additional heterozygous and wild-type

Results and discussion

To determine if the route of injection significantly impacts the degree to which the SMA phenotype is corrected in SMA mice following treatment with scAAV9-SMN, we performed a titration experiment to identify a relatively low concentration of vector that still resulted in significant survival (data not shown). This concentration (2 × 1010) as opposed to a maximal dose was used in subsequent experiments since a lower concentration was more likely to reveal differences regarding the ability to

Acknowledgments

We thank John R. Marston for expert technical assistance and Hansjörg Rindt, PhD for his assistance in statistical analysis. This work was supported by Grants from the National Institutes of Health [R01 HD054413; R01 HS41584], a training fellowship for JJG [NIGMS T32] and a Grant from SMA Europe (MS).

References (24)

  • T.O. Crawford et al.

    The neurobiology of childhood spinal muscular

    Neurobiol. Dis.

    (1996)
  • S. Lefebvre et al.

    Identification and characterization of a spinal muscular atrophy-determining gene

    Cell

    (1995)
  • M.E. Butchbach et al.

    Abnormal motor phenotype in the SMNDelta7 mouse model of spinal muscular atrophy

    Neurobiol. Dis.

    (2007)
  • M. Oskoui et al.

    The changing natural history of spinal muscular atrophy type 1

    Neurology

    (2007)
  • C.F. Rochette et al.

    SMN gene duplication and the emergence of the SMN2 gene occurred in distinct hominids: SMN2 is unique to Homo sapiens

    Hum. Genet.

    (2001)
  • C.L. Lorson et al.

    A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy

    Proc. Natl. Acad. Sci. USA

    (1999)
  • L. Cartegni et al.

    Disruption of an SF2/ASF-dependent exonic splicing enhancer in SMN2 causes spinal muscular atrophy in the absence of SMN1

    Nat. Genet.

    (2002)
  • T.T. Le et al.

    SMNDelta7, the major product of the centromeric survival motor neuron (SMN2) gene, extends survival in mice with spinal muscular atrophy and associates with full-length SMN

    Hum. Mol. Genet.

    (2005)
  • M.A. Passini et al.

    CNS-targeted gene therapy improves survival and motor function in a mouse model of spinal muscular atrophy

    J. Clin. Invest.

    (2010)
  • K.D. Foust et al.

    Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN

    Nat. Biotechnol.

    (2010)
  • E. Dominguez et al.

    Intravenous scAAV9 delivery of a codon-optimized SMN1 sequence rescues SMA mice

    Hum. Mol. Genet.

    (2011)
  • C.F. Valori et al.

    Systemic delivery of scAAV9 expressing SMN prolongs survival in a model of spinal muscular atrophy

    Sci. Transl. Med.

    (2010)
  • Cited by (56)

    • Challenges of gene delivery to the central nervous system and the growing use of biomaterial vectors

      2019, Brain Research Bulletin
      Citation Excerpt :

      Spinal muscular atrophy is a genetic neurodegenerative disorder where α-motor neurons are lost, leading to progressive skeletal muscle atrophy (D’Amico et al., 2011). Several researchers show that delivering genes encoding functional survival motor neuron protein (SMN) can effectively treat spinal muscular atrophy (Benkhelifa-Ziyyat et al., 2013; Dominguez et al., 2011; Foust et al., 2010; Glascock et al., 2012; Valori et al., 2010). Several of these studies administered the vector peripherally and, therefore, the injection required a high concentration of vector system to produce an efficacious effect.

    • Spinal Muscular Atrophy Therapeutics Development

      2017, Spinal Muscular Atrophy: Disease Mechanisms and Therapy
    • The retinoid agonist tazarotene promotes angiogenesis and wound healing

      2016, Molecular Therapy
      Citation Excerpt :

      The growth of new functional blood vessel network is determined by the extent of branching and tubulogenesis. The endothelial tube formation assay recapitulates most cellular process involves in the assembly of endothelial cells into a primary plexus (e.g., cell–cell, cell–matrix interaction) and has a high predictive value for angiogenesis-related cellular morphogenesis such as vascular branching and, suggesting that this is an appropriate model for in vivo angiogenesis.11,12 We developed a high content imaging approach using the endothelial tube formation assay to screen small molecule libraries with aim of identifying novel therapeutic molecules that enhance new blood vessel growth.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text