Original article
Partial restoration of cardio-vascular defects in a rescued severe model of spinal muscular atrophy

https://doi.org/10.1016/j.yjmcc.2012.01.005Get rights and content

Abstract

Spinal muscular atrophy (SMA) is a leading genetic cause of infantile death. Loss of a gene called Survival Motor Neuron 1 (SMN1) and, as a result, reduced levels of the Survival Motor Neuron (SMN) protein leads to SMA development. SMA is characterized by the loss of functional motor neurons in the spinal cord. However, accumulating evidence suggests the contribution of other organs to the composite SMA phenotype and disease progression. A growing number of congenital heart defects have been identified in severe SMA patients. Consistent with the clinical cases, we have recently identified developmental and functional heart defects in two SMA mouse models, occurring at embryonic stage in a severe SMA model and shortly after birth in a less severe model (SMN∆7). Our goal was to examine the late stage cardiac abnormalities in untreated SMN∆7 mice and to determine whether gene replacement therapy restores cardiac structure/function in rescued SMN∆7 model. To reveal the extent of the cardiac structural/functional repair in the rescued mice, we analyzed the heart of untreated and treated SMN∆7 model using self-complementary Adeno-associated virus (serotype 9) expressing the full-length SMN cDNA. We examined the characteristics of the heart failure such as remodeling, fibrosis, oxidative stress, and vascular integrity in both groups. Our results clearly indicate that fibrosis, oxidative stress activation, vascular remodeling, and a significant decrease in the number of capillaries exist in the SMA heart. The cardiac structural defects were improved drastically in the rescued animals, however, the level of impairment was still significant compared to the age-matched wildtype littermates. Furthermore, functional analysis by in vivo cardiac magnetic resonance imaging (MRI) revealed that the heart of the treated SMA mice still exhibits functional defects. In conclusion, cardiac abnormalities are only partially rescued in post-birth treated SMA animals and these abnormalities may contribute to the premature death of vector-treated SMA animals with seemingly rescued motor function but an average life span of less than 70 days as reported in several studies.

Highlights

► Rescued SMA mice demonstrate improvements in cardiac structural and cellular pathology. ► Vascular and capillary defects are improved in rescued mice. ► Cine MRI indicates cardiac functional impairments in rescued SMA mice. ► Functional defects are most likely contributing to the premature death of treated SMA mice.

Introduction

Spinal muscular atrophy (SMA) is the leading genetic cause of infantile death and is characterized by the loss of the α-motor neurons in the spinal cord [1]. The clinical spectrum is broadly categorized based upon disease onset and physical milestones [2]. SMA results from the loss of survival motor neuron-1 (SMN1) [3]. A human-specific copy gene is present on the same chromosome called SMN2 [3], [4]. SMN2 is nearly identical to SMN1 and generates low levels of full-length SMN. SMN2 cannot prevent disease development in the absence of SMN1 since the majority of SMN2-derived transcripts are alternatively spliced and encode an unstable protein [5], [6].

The fundamental pathology in SMA is neurodegeneration, however, there are clinical reports demonstrating the contribution of other tissues to the overall phenotype in the most severe forms (Type 0 and Type I). In severe SMA, several congenital heart defects have been documented including atrial septal defects, dilated right ventricle, ventricular septal defects, and hypoplastic left heart syndrome [[7], [8], [9]]. Sudden death without a clear explanation can occur in severe SMA patients [10]. Central apnea due to bulbar dysfunction and respiratory complications is suggested to be the possible cause of sudden death, even though it also occurs in patients with ventilation support [10]. Therefore, vagal hypertonia induced bradycardia has also been considered [10]. Furthermore, palpitations, ST-segment abnormalities, couplets, diastolic dysfunction, and right ventricular overload have been reported in SMA patients [11], [12]. Distal necrosis has also been documented in severe SMA, occasionally with atrial septal defects and asymmetric left ventricular hypertrophy [13], [14]. Similar pathologies have been observed in SMA mice treated with therapeutic compounds or viral vectors [15], [16]. Life span extension of treated SMA mice has led to increased recognition of distal necrosis in the tail or ear pinna. These results suggest that the dysfunction of the autonomic nervous system may lead to impaired regulation of vascular tone and possibly fetal vasculature defects which cannot be completely restored with post-birth treatments. Recently, our lab and others have identified functional and structural heart defects in SMA animal models [[17], [18], [19]]. Bradycardia is the most common autonomic nervous system phenotype reported in these studies. We have identified cardiac defects occurring during embryonic development in a severe SMA mouse model and shortly after birth in a slightly less severe model (SMNΔ7), suggesting that cardiac defects precede motor neuron degeneration.

Recently, intravenous delivery at postnatal day 1 (P1) of scAAV9 expressing full-length SMN cDNA in SMNΔ7 mice led to a profound phenotypic rescue by restoring motor function/neuromuscular physiology and extended survival up to 250 days [20]. The same group reported complete restoration of the heart rate and partial repair of cardiac function in AAV9-injected animals at P14 [17]. Additional groups reported a significantly extended life span using a similar vector, however, sudden deaths, respiratory complications, and average life spans of only 65–70 days were common [21], [22]. To provide a more detailed analysis of the cardiac defects in these important disease models, we have examined remodeling, fibrosis, oxidative stress, and vascular impairments. In each instance, the SMA mice were profoundly impaired compared to healthy animals and these results served as an important baseline from which we could quantitatively assess cardiac structure/function following SMN gene replacement. To directly compare the functional parameters, we carried out in vivo MRI on the heart of the rescued and age-matched wildtype littermates. Our study demonstrates that the heart of the rescued SMA mice, albeit drastically improved compared to untreated mice, still exhibits structural and functional defects compared to age-matched wildtype mice, suggesting that cardiac defects contribute to the shortened life span of scAAV-rescued mice [21], [22].

Section snippets

SMA animals and injection

All animal experiments took place in accordance with procedures approved by NIH guidelines and MU Animal Care and Use Committee (ACUC). SMNΔ7 animals (mSmn −/−, hSMN2+/+, SMNΔ7+/+) were genotyped at P1 [23]. scAAV9 expressing the full-length SMN cDNA was purified by CsCl centrifugation [20] and SMNΔ7 P2 mice were injected intravenously into the temporal facial vein with 1 × 1011 viral genomes.

Histology

Animals were anesthetized using 0.06 ml/g isoflurane (Vet One) for 3–5 min (adequacy of anesthesia was

Gene replacement therapy rescues cardiac remodeling

A widely utilized SMA mouse model is referred to “SMNΔ7”. SMNΔ7 model lacks murine Smn, contains two copies of human SMN2, and includes an additional transgene expressing SMNΔ7 cDNA (mSmn−/−, hSMN2+/+, SMNΔ7+/+) with a life span of 14–16 days [29]. To obtain rescued SMA mice, 1 × 1011 genomic copies of scAAV9-SMN, a vector expressing the full-length SMN cDNA, was intravenously (IV) injected into SMNΔ7 mice. This delivery mode allows distribution of the virus to the central nervous system as well

Discussion

Gene replacement with scAAV-SMN has provided the most substantial correction in severe SMA mice [[15], [20], [21], [22]]. However, there were intriguing distinctions regarding the survival rate between the various studies. Interestingly, SMN delivered by scAAV8 via intracerebroventricular injection led to a partial phenotypic rescue with a shorter average life span than systemic injection of scAAV9-SMN [15], [20]. This difference may be due to: 1) the ability of scAAV9 to provide global cardiac

Funding

This work was supported by grants from the National Institutes of Health, (R01 HL107910-01, R01 HL107910-01 to J.R.S.), a Faculty Research Award from the MU College of Veterinary Medicine to C.L.L. and SMA Europe to M.S.

The following are the supplementary materials related to this article.

Conflict of interest statement

None declared.

Acknowledgment

We would like to thank members of the Lorson lab for their helpful discussions and Dr. Hans Rindt for the generous gift of WGA. In vivo MRI was performed at the Biomolecular Imaging Center supported by the Harry S. Truman Veterans Affairs Hospital at the University of Missouri and we greatly appreciate the technical assistance from Ming Yang.

References (55)

  • W.M. Mulleners et al.

    Spinal muscular atrophy combined with congenital heart disease: a report of two cases

    Neuropediatrics

    (1996)
  • S. Rudnik-Schoneborn et al.

    Congenital heart disease is a feature of severe infantile spinal muscular atrophy

    J Med Genet

    (2008)
  • P. Moller et al.

    Spinal muscular atrophy type i combined with atrial septal defect in three sibs

    Clin Genet

    (1990)
  • J. Finsterer et al.

    Cardiac involvement in Werdnig–Hoffmann's spinal muscular atrophy

    Cardiology

    (1999)
  • G. Distefano et al.

    Heart involvement in progressive spinal muscular atrophy. A review of the literature and case histories in childhood

    Pediatr Med Chir

    (1994)
  • S. Rudnik-Schoneborn et al.

    Digital necroses and vascular thrombosis in severe spinal muscular atrophy

    Muscle Nerve

    (2010)
  • Q. Araujo Ade et al.

    Vascular perfusion abnormalities in infants with spinal muscular atrophy

    J Pediatr

    (2009)
  • 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)
  • A.M. Avila et al.

    Trichostatin A increases SMN expression and survival in a mouse model of spinal muscular atrophy

    J Clin Invest

    (2007)
  • A.K. Bevan et al.

    Early heart failure in the SMN{delta}7 model of spinal muscular atrophy and correction by postnatal scAAV9-smn delivery

    Hum Mol Genet

    (2010)
  • C.R. Heier et al.

    Arrhythmia and cardiac defects are a feature of spinal muscular atrophy model mice

    Hum Mol Genet

    (2010)
  • M. Shababi et al.

    Cardiac defects contribute to the pathology of spinal muscular atrophy models

    Hum Mol Genet

    (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)
  • C.F. Valori et al.

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

    Sci Transl Med

    (2010)
  • E. Dominguez et al.

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

    Hum Mol Genet

    (2011)
  • T.H. Coady et al.

    Development of a single vector system that enhances trans-splicing of SMN2 transcripts

    PLoS One

    (2008)
  • M. Shababi et al.

    Combination of SMN trans-splicing and a neurotrophic factor increases the life span and body mass in a severe model of spinal muscular atrophy

    Hum Gene Ther

    (2010)
  • Cited by (55)

    • Spinal muscular atrophy: Broad disease spectrum and sex-specific phenotypes

      2021, Biochimica et Biophysica Acta - Molecular Basis of Disease
    • AAV9-DOK7 gene therapy reduces disease severity in Smn<sup>2B/-</sup> SMA model mice

      2020, Biochemical and Biophysical Research Communications
    • Motor transmission defects with sex differences in a new mouse model of mild spinal muscular atrophy

      2020, EBioMedicine
      Citation Excerpt :

      Over the past 10 years, a resurgence of case studies has highlighted potential heart defects in severe SMA patients [40]. Similarly, multiple studies of the heart in preclinical models demonstrated bradycardia, reduced function, innervation and vascularization [41-44]. We investigated whether cardiac defects were present in 12 and 18 month old Smn2B/−;SMN2+/- mice using echocardiogram.

    • Into the unknown: Chromatin signaling in spinal muscular atrophy

      2019, Chromatin Signaling and Neurological Disorders
    • A Direct Comparison of IV and ICV Delivery Methods for Gene Replacement Therapy in a Mouse Model of SMARD1

      2018, Molecular Therapy Methods and Clinical Development
      Citation Excerpt :

      Qualitative and quantitative measurements were analyzed offline using the Vevo software. 100 mg spinal cord and heart tissue were homogenized in 200–500 μL Jurkat lysis buffer (JLB) and equal amounts of protein were loaded onto 10% SDS-PAGE gel as previously described.37,40,41 Blots were incubated overnight at 4°C with rabbit IGHMBP2 antibody (Aviva Systems Biology, San Diego, CA) diluted to 1:1,000, and horseradish peroxidase (HRP) anti-rabbit immunoglobulin G (IgG) (Jackson Immunoresearch) was used as secondary antibody.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text