Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

AAV-based gene therapy prevents neuropathology and results in normal cognitive development in the hyperargininemic mouse

Abstract

Complete arginase I deficiency is the least severe urea cycle disorder, characterized by hyperargininemia and infrequent episodes of hyperammonemia. Patients suffer from neurological impairment with cortical and pyramidal tract deterioration, spasticity, loss of ambulation and seizures, and is associated with intellectual disability. In mice, onset is heralded by weight loss beginning around day 15; gait instability follows progressing to inability to stand and development of tail tremor with seizure-like activity and death. Here we report that hyperargininemic mice treated neonatally with an adeno-associated virus (AAV)-expressing arginase and followed long-term lack any presentation consistent with brain dysfunction. Behavioral and histopathological evaluation demonstrated that treated mice are indistinguishable from littermates, and that putative compounds associated with neurotoxicity are diminished. In addition, treatment results in near complete resolution of metabolic abnormalities early in life; however, there is the development of some derangement later with decline in transgene expression. Ammonium challenging revealed that treated mice are affected by exogenous loading much greater than littermates. These results demonstrate that AAV-based therapy for hyperargininemia is effective and prevents development of neurological abnormalities and cognitive dysfunction in a mouse model of hyperargininemia; however, nitrogen challenging reveals that these mice remain impaired in the handling of waste nitrogen.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7
Figure 8
Figure 9
Figure 10

Similar content being viewed by others

References

  1. Brusilow SW, Horwich A . The metabolic and molecular bases of inherited disease. In: Scriver CR, Beaudet AL, Sly WS, Valle D (eds) The metabolic and Molecular Bases of Inherited Disease 8th edn. McGraw-Hill: New York pp 1909–1963 2001).

    Google Scholar 

  2. Iyer R, Jenkinson CP, Vockley JG, Kern RM, Grody WW, Cederbaum S . The human arginases and arginase deficiency. J Inherit Metab Dis 1998; 21: 86–100.

    Article  CAS  Google Scholar 

  3. Jain-Ghai S, Nagamani SC, Blaser S, Siriwardena K, Feigenbaum A . Arginase I deficiency: severe infantile presentation with hyperammonemia: more common than reported? Mol Genet Metab 2011; 104: 107–111.

    Article  CAS  Google Scholar 

  4. Picker JD, Puga AC, Levy HL, Marsden D, Shih VE, Degirolami U et al. Arginase deficiency with lethal neonatal expression: evidence for the glutamine hypothesis of cerebral edema. J Pediatr 2003; 142: 349–352.

    Article  Google Scholar 

  5. Prasad AN, Breen JC, Ampola MG, Rosman NP . Argininemia: a treatable genetic cause of progressive spastic diplegia simulating cerebral palsy: case reports and literature review. J Child Neurol 1997; 12: 301–309.

    Article  CAS  Google Scholar 

  6. Iyer RK, Yoo PK, Kern RM, Rozengurt N, Tsoa R, O’Brien WE et al. Mouse model for human arginase deficiency. Mol Cell Biol 2002; 22: 4491–4498.

    Article  CAS  Google Scholar 

  7. Lee EK, Hu C, Bhargava R, Rozengurt N, Stout D, Grody WW et al. Long-term survival of the juvenile lethal arginase deficient mouse with aav gene therapy. Mol Ther 2012; 20: 1844–1851.

    Article  CAS  Google Scholar 

  8. Hu C, Busuttil RW, Lipshutz GS . RH10 provides superior transgene expression in mice when compared with natural AAV serotypes for neonatal gene therapy. J Gene Med 2010; 12: 766–778.

    Article  CAS  Google Scholar 

  9. Hu C, Lipshutz GS . AAV-based neonatal gene therapy for hemophilia A: long-term correction and avoidance of immune responses in mice. Gene Therapy 2012; 19: 1166–1176.

    Article  CAS  Google Scholar 

  10. Wolfer DP, Stagljar-Bozicevic M, Errington ML, Lipp HP . Spatial memory and learning in transgenic mice: fact or artifact? News Physiol Sci 1998; 13: 118–123.

    PubMed  Google Scholar 

  11. Clapcote SJ, Lazar NL, Bechard AR, Wood GA, Roder JC . NIH Swiss and Black Swiss mice have retinal degeneration and performance deficits in cognitive tests. Comp Med 2005; 55: 310–316.

    CAS  PubMed  Google Scholar 

  12. Jenkinson CP, Grody WW, Cederbaum SD . Comparative properties of arginases. Comp Biochem Physiol B Biochem Mol Biol 1996; 114: 107–132.

    Article  CAS  Google Scholar 

  13. Singh R, Pervin S, Karimi A, Cederbaum S, Chaudhuri G . Arginase activity in human breast cancer cell lines: N(omega)-hydroxy-L-arginine selectively inhibits cell proliferation and induces apoptosis in MDA-MB-468 cells. Cancer Res 2000; 60: 3305–3312.

    CAS  PubMed  Google Scholar 

  14. Marescau B, Qureshi IA, De Deyn P, Letarte J, Ryba R, Lowenthal A . Guanidino compounds in plasma, urine and cerebrospinal fluid of hyperargininemic patients during therapy. Clin Chim Acta 1985; 146: 21–27.

    Article  CAS  Google Scholar 

  15. Marescau B, De Deyn PP, Lowenthal A, Qureshi IA, Antonozzi I, Bachmann C et al. Guanidino compound analysis as a complementary diagnostic parameter for hyperargininemia: follow-up of guanidino compound levels during therapy. Pediatr Res 1990; 27: 297–303.

    Article  CAS  Google Scholar 

  16. Mizutani N, Hayakawa C, Ohya Y, Watanabe K, Watanabe Y, Mori A . Guanidino compounds in hyperargininemia. Tohoku J Exp Med 1987; 153: 197–205.

    Article  CAS  Google Scholar 

  17. Terheggen HG, Lavinha F, Colombo JP, Van Sande M, Lowenthal A . Familial hyperargininemia. J Genet Hum 1972; 20: 69–84.

    CAS  PubMed  Google Scholar 

  18. Wiechert P, Mortelmans J, Lavinha F, Clara R, Terheggen HG, Lowenthal A . Excretion of guanidino-derivates in urine of hyperargininemic patients. J Genet Hum 1976; 24: 61–72.

    CAS  PubMed  Google Scholar 

  19. De Deyn PP, Marescau B, Macdonald RL . Effects of alpha-keto-delta-guanidinovaleric acid on inhibitory amino acid responses on mouse neurons in cell culture. Brain Res 1988; 449: 54–60.

    Article  CAS  Google Scholar 

  20. De Deyn PP, Marescau B, Macdonald RL . Guanidino compounds that are increased in hyperargininemia inhibit GABA and glycine responses on mouse neurons in cell culture. Epilepsy Res 1991; 8: 134–141.

    Article  CAS  Google Scholar 

  21. Balz D, de Souza Wyse AT, Morsch VM, da Silva AC, Vieira VL, Morsch AL et al. In vitro effects of L-arginine and guanidino compounds on NTPDase1 and 5′-nucleotidase activities from rat brain synaptosomes. Int J Dev Neurosci 2003; 21: 75–82.

    Article  CAS  Google Scholar 

  22. Deignan JL, De Deyn PP, Cederbaum SD, Fuchshuber A, Roth B, Gsell W et al. Guanidino compound levels in blood, cerebrospinal fluid, and post-mortem brain material of patients with argininemia. Mol Genet Metab 2010; 100: S31–S36.

    Article  CAS  Google Scholar 

  23. Marescau B, De Deyn PP, Qureshi IA, De Broe ME, Antonozzi I, Cederbaum SD et al. The pathobiochemistry of uremia and hyperargininemia further demonstrates a metabolic relationship between urea and guanidinosuccinic acid. Metabolism 1992; 41: 1021–1024.

    Article  CAS  Google Scholar 

  24. Marescau B, Nagels G, Possemiers I, De Broe ME, Becaus I, Billiouw JM et al. Guanidino compounds in serum and urine of nondialyzed patients with chronic renal insufficiency. Metabolism 1997; 46: 1024–1031.

    Article  CAS  Google Scholar 

  25. Ceriotti G . Ultramicrodetermination of plasma urea by reaction with diacetylmonoxime--antipyrine without deproteinization. Clin Chem 1971; 17: 400–402.

    CAS  PubMed  Google Scholar 

  26. Wang T, Lawler AM, Steel G, Sipila I, Milam AH, Valle D . Mice lacking ornithine-delta-aminotransferase have paradoxical neonatal hypoornithinaemia and retinal degeneration. Nat Genet 1995; 11: 185–190.

    Article  Google Scholar 

  27. Rogers DC, Fisher EM, Brown SD, Peters J, Hunter AJ, Martin JE . Behavioral and functional analysis of mouse phenotype: SHIRPA, a proposed protocol for comprehensive phenotype assessment. Mamm Genome 1997; 8: 711–713.

    Article  CAS  Google Scholar 

  28. Irwin S . Comprehensive observational assessment: Ia. A systematic, quantitative procedure for assessing the behavioral and physiologic state of the mouse. Psychopharmacologia 1968; 13: 222–257.

    Article  CAS  Google Scholar 

  29. Rafael JA, Nitta Y, Peters J, Davies KE . Testing of SHIRPA, a mouse phenotypic assessment protocol, on Dmd(mdx) and Dmd(mdx3cv) dystrophin-deficient mice. Mamm Genome 2000; 11: 725–728.

    Article  CAS  Google Scholar 

  30. Hogg S . A review of the validity and variability of the elevated plus-maze as an animal model of anxiety. Pharmacol Biochem Behav 1996; 54: 21–30.

    Article  CAS  Google Scholar 

  31. Walf AA, Frye CA . The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nat Protoc 2007; 2: 322–328.

    Article  CAS  Google Scholar 

  32. Vorhees CV, Williams MT . Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nat Protoc 2006; 1: 848–858.

    Article  Google Scholar 

  33. Bryant CD, Eitan S, Sinchak K, Fanselow MS, Evans CJ . NMDA receptor antagonism disrupts the development of morphine analgesic tolerance in male, but not female C57BL/6J mice. Am J Physiol Regul Integr Comp Physiol 2006; 291: R315–R326.

    Article  CAS  Google Scholar 

  34. Stout DB, Chatziioannou AF, Lawson TP, Silverman RW, Gambhir SS, Phelps ME . Small animal imaging center design: the facility at the UCLA Crump Institute for Molecular Imaging. Mol Imaging Biol 2005; 7: 393–402.

    Article  Google Scholar 

  35. Ye X, Robinson MB, Pabin C, Quinn T, Jawad A, Wilson JM et al. Adenovirus-mediated in vivo gene transfer rapidly protects ornithine transcarbamylase-deficient mice from an ammonium challenge. Pediatr Res 1997; 41 (4 Pt 1): 527–534.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Waldemar Ladno for assistance with CT imaging of mice and Daniela Markovic for assistance with the statistical evaluation, and both the Semel Institute for Neuroscience and the Intellectual and Developmental Disabilities Research Center at UCLA for their support. The authors declare no financial or other conflict of interest. This work was supported by grants from the NIH (5K08HD057555-05 and 1R01NS071076-02A1).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to G S Lipshutz.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies the paper on Gene Therapy website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lee, E., Hu, C., Bhargava, R. et al. AAV-based gene therapy prevents neuropathology and results in normal cognitive development in the hyperargininemic mouse. Gene Ther 20, 785–796 (2013). https://doi.org/10.1038/gt.2012.99

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/gt.2012.99

Keywords

This article is cited by

Search

Quick links