J Korean Neurosurg Soc.  2020 Sep;63(5):579-589. 10.3340/jkns.2019.0182.

An Optimization of AAV-82Q-Delivered Rat Model of Huntington’s Disease

Affiliations
  • 1Institute for Stem Cell & Regenerative Medicine (ISCRM), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
  • 2Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
  • 3Department of Neurosurgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 4Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea
  • 5Department of Neurosurgery, Chungbuk National University Hospital, Cheongju, Korea
  • 6Department of Biochemistry and Medical Research Center, Chungbuk National University, Cheongju, Korea
  • 7Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea

Abstract


Objective
: No optimum genetic rat Huntington model both neuropathological using an adeno-associated virus (AAV-2) vector vector has been reported to date. We investigated whether direct infection of an AAV2 encoding a fragment of mutant huntingtin (AV2-82Q) into the rat striatum was useful for optimizing the Huntington rat model.
Methods
: We prepared ten unilateral models by injecting AAV2-82Q into the right striatum, as well as ten bilateral models. In each group, five rats were assigned to either the 2×1012 genome copies (GC)/mL of AAV2-82Q (×1, low dose) or 2×1013 GC/mL of AAV2-82Q (×10, high dose) injection model. Ten unilateral and ten bilateral models injected with AAV-empty were also prepared as control groups. We performed cylinder and stepping tests 2, 4, 6, and 8 weeks after injection, tested EM48 positive mutant huntingtin aggregates.
Results
: The high dose of unilateral and bilateral AAV2-82Q model showed a greater decrease in performance on the stepping and cylinder tests. We also observed more prominent EM48-positive mutant huntingtin aggregates in the medium spiny neurons of the high dose of AAV2-82Q injected group.
Conclusion
: Based on the results from the present study, high dose of AAV2-82Q is the optimum titer for establishing a Huntington rat model. Delivery of high dose of human AAV2-82Q resulted in the manifestation of Huntington behaviors and optimum expression of the huntingtin protein in vivo.

Keyword

Huntington disease; Adeno-associated virus vector; Huntingtin protein; Neurodegenerative diseases; Gene delivery

Figure

  • Fig. 1. Schematic of the study protocol. AAV2 : adeno-associated viral vector, CPU : caudate putamen, MRI : magnetic resonance imaging.

  • Fig. 2. The number of steps in the stepping test recorded at 2, 4, 6, and 8 weeks after the AAV2 injection. A : The use of the left paw by the ×1 and ×10 subgroups of the unilateral group as significantly decreased compared with the control subgroup after two weeks. Additionally, the use of the left paw was significantly decreased over time in both the ×1 and ×10 subgroups. At week 8, a significant difference was observed between the ×1 and ×10 subgroups. B : No differences were observed among the three subgroups. C and D : The use of both paws was significantly decreased in the ×1 and ×10 subgroups of the bilateral group compared to the control subgroup after 4 weeks. At week 8, a significant difference was observed between the ×1 and ×10 subgroups. Additionally, the use of both paws was significantly decreased over time in both the ×1 and ×10 subgroups. *p<0.001. † p<0.01. ‡ p<0.05.

  • Fig. 3. The asymmetry scores for the cylinder test at 2, 4, 6, and 8 weeks after the AAV2 injection. A : The asymmetry scores of the ×1 and ×10 subgroups of the unilateral group were significantly decreased compared with the control subgroup at 6 and 8 weeks. B : The asymmetry scores did not differ among the three subgroups of the bilateral group. *p<0.001.

  • Fig. 4. Immunohistochemical staining with the EM48 antibody showed aggregation of the hTT protein in the striatum of the hD rats. A and B : Formation of mutant hTT detected by the EM48 antibody at 11 weeks after AAV2 injection into the contralateral and ipsilateral striatum of the hD rats. Arrows indicate intranuclear aggregates, and arrowheads indicate smaller neuropil aggregates outside the nucleus. C and D : The number of EM48 aggregated cells counted was significantly different between the hD ×1 and hD ×10 subgroups in unilateral ipsilateral striatum (C) and bilateral striata (D) (one-way ANOVA with Turkey’s post-hoc test). *p<0.05. hTT : huntingtin gene, hD : huntington’s disease, AAV2 : adeno-associated viral vector, ANOVA : analysis of variance.

  • Fig. 5. The huntingtin gene (hTT) protein expression was detected by an hTT-specific antibody (mAB2166) in the striatum of AAV-82Q-infected rat brain. The mAb2166 positive cells (cells with brown color) were found with higher expression in the AAV2-82Q ×10 infected subgroup (D and h) than in the x1 subgroup (C and G). The mAb2166 was scarcely positive in the control subgroups (A, B, E, and F). Scale bar, A-D : 50 μm; E-h : 20 μm. Brian tissue was embedded in paraffin, and hematoxyin was used. A-D are 10× image and E-h are 20× image. hD : huntington’s disease, AAV2 : adeno-associated virus.

  • Fig. 6. The histological characters of the AAV-82Q huntingtin infected rat brain. Brain sections showed the activation of microglia and astrocytes in AAV-82Q rats. higher expression of an oligodendrocyte marker (Olig2) and an apoptosis marker (Caspase3) were observed in the AAV2-82Q ×10 group compared to control groups and the ×1 group. The neuronal cell marker (NeuN) was not different between control and injected groups. Scale bar : 50 μm. hD : huntington’s disease, AAV2 : adeno-associated virus.


Reference

References

1. Alexander GE, Crutcher MD. Preparation for movement: neural representations of intended direction in three motor areas of the monkey. J Neurophysiol. 64:133–150. 1990.
Article
2. Cattoglio C, Facchini G, Sartori D, Antonelli A, Miccio A, Cassani B, et al. Hot spots of retroviral integration in human CD34+ hematopoietic cells. Blood. 110:1770–1778. 2007.
Article
3. Ceccarelli I, Fiengo P, Remelli R, Miragliotta V, Rossini L, Biotti I, et al. Recombinant Adeno Associated Viral (AAV) vector type 9 delivery of Ex1-Q138-mutant huntingtin in the rat striatum as a short-time model for in vivo studies in drug discovery. Neurobiol Dis. 86:41–51. 2016.
Article
4. Davies SW, Turmaine M, Cozens BA, DiFiglia M, Sharp AH, Ross CA, et al. Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation. Cell. 90:537–548. 1997.
Article
5. Daya S, Berns KI. Gene therapy using adeno-associated virus vectors. Clin Microbiol Rev. 21:583–593. 2008.
Article
6. de Almeida LP, Ross CA, Zala D, Aebischer P, Déglon N. Lentiviralmediated delivery of mutant huntingtin in the striatum of rats induces a selective neuropathology modulated by polyglutamine repeat size, huntingtin expression levels, and protein length. J Neurosci. 22:3473–3483. 2002.
Article
7. DiFiglia M, Sapp E, Chase KO, Davies SW, Bates GP, Vonsattel JP, et al. Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. Science. 277:1990–1993. 1997.
Article
8. Foster AC, Whetsell WO Jr, Bird ED, Schwarcz R. Quinolinic acid phosphoribosyltransferase in human and rat brain: activity in Huntington’s disease and in quinolinate-lesioned rat striatum. Brain Res. 336:207–214. 1985.
Article
9. Hodgson JG, Agopyan N, Gutekunst CA, Leavitt BR, LePiane F, Singaraja R, et al. A YAC mouse model for Huntington’s disease with full-length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurode generation. Neuron. 23:181–192. 1999.
Article
10. Hoth KF, Paulsen JS, Moser DJ, Tranel D, Clark LA, Bechara A. Patients with Huntington’s disease have impaired awareness of cognitive, emotional, and functional abilities. J Clin Exp Neuropsychol. 29:365–376. 2007.
Article
11. Hua Y, Schallert T, Keep RF, Wu J, Hoff JT, Xi G. Behavioral tests after intracerebral hemorrhage in the rat. Stroke. 33:2478–2484. 2002.
Article
12. Jang M, Lee SE, Cho IH. Adeno-associated viral vector serotype DJmediated overexpression of N171-82Q-mutant huntingtin in the striatum of juvenile mice is a new model for Huntington’s disease. Front Cell Neurosci. 12:157. 2018.
Article
13. Kim M. Brain Transplantation for Huntington Disease. Tissue Eng Regen Med. 2:322–326. 2005.
14. MacDonald ME, Ambrose CM, Duyao MP, Myers RH, Lin C, Srinidhi L et al. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. The Huntington’s Disease Collaborative Research Group. Cell. 72:971–983. 1993.
Article
15. Mangiarini L, Sathasivam K, Mahal A, Mott R, Seller M, Bates GP. Instability of highly expanded CAG repeats in mice transgenic for the Huntington’s disease mutation. Nat Genet. 15:197–200. 1997.
Article
16. Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C, et al. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell. 87:493–506. 1996.
Article
17. Marini B, Kertesz-Farkas A, Ali H, Lucic B, Lisek K, Manganaro L, et al. Nuclear architecture dictates HIV-1 integration site selection. Nature. 521:227–231. 2015.
Article
18. Nussbaum RL, McInnes RR, Willard HF. Thompson & Thompson genetics in medicine. Philadelphia: Elsevier Health Sciences;2015.
19. Olsson M, Nikkhah G, Bentlage C, Björklund A. Forelimb akinesia in the rat Parkinson model: differential effects of dopamine agonists and nigral transplants as assessed by a new stepping test. J Neurosci. 15(5 Pt 2):3863–3875. 1995.
Article
20. Palfi S, Brouillet E, Jarraya B, Bloch J, Jan C, Shin M, et al. Expression of mutated huntingtin fragment in the putamen is sufficient to produce abnormal movement in non-human primates. Mol Ther. 15:1444–1451. 2007.
Article
21. Radua J, van den Heuvel OA, Surguladze S, Mataix-Cols D. Metaanalytical comparison of voxel-based morphometry studies in obsessivecompulsive disorder vs other anxiety disorders. Arch Gen Psychiatry. 67:701–711. 2010.
Article
22. Ramaswamy S, McBride JL, Kordower JH. Animal models of Huntington’s disease. ILAR J. 48:356–373. 2007.
Article
23. Reddy PH, Charles V, Williams M, Miller G, Whetsell WO Jr, Tagle DA. Transgenic mice expressing mutated full-length HD cDNA: a paradigm for locomotor changes and selective neuronal loss in Huntington’s disease. Philos Trans R Soc Lond B Biol Sci. 354:1035–1045. 1999.
Article
24. Reddy PH, Williams M, Charles V, Garrett L, Pike-Buchanan L, Whetsell WO Jr, et al. Behavioural abnormalities and selective neuronal loss in HD transgenic mice expressing mutated full-length HD cDNA. Nat Genet. 20:198–202. 1998.
Article
25. Reddy PH, Williams M, Tagle DA. Recent advances in understanding the pathogenesis of Huntington’s disease. Trends Neurosci. 22:248–255. 1999.
Article
26. Régulier E, Trottier Y, Perrin V, Aebischer P, Déglon N. Early and reversible neuropathology induced by tetracycline-regulated lentiviral overexpression of mutant huntingtin in rat striatum. Hum Mol Genet. 12:2827–2836. 2003.
Article
27. Sanberg PR, Calderon SF, Giordano M, Tew JM, Norman AB. The quinolinic acid model of Huntington’s disease: locomotor abnormalities. Exp Neurol. 105:45–53. 1989.
Article
28. Schilling G, Becher MW, Sharp AH, Jinnah HA, Duan K, Kotzuk JA, et al. Intranuclear inclusions and neuritic aggregates in transgenic mice expressing a mutant N-terminal fragment of huntingtin. Hum Mol Genet. 8:397–407. 1999.
Article
29. Senut MC, Suhr ST, Kaspar B, Gage FH. Intraneuronal aggregate formation and cell death after viral expression of expanded polyglutamine tracts in the adult rat brain. J Neurosci. 20:219–229. 2000.
Article
30. Shelbourne PF, Killeen N, Hevner RF, Johnston HM, Tecott L, Lewandoski M, et al. A Huntington’s disease CAG expansion at the murine Hdh locus is unstable and associated with behavioural abnormalities in mice. Hum Mol Genet. 8:763–774. 1999.
Article
31. Slow EJ, van Raamsdonk J, Rogers D, Coleman SH, Graham RK, Deng Y, et al. Selective striatal neuronal loss in a YAC128 mouse model of Huntington disease. Hum Mol Genet. 12:1555–1567. 2003.
Article
32. Stack EC, Ferrante RJ. Huntington’s disease: progress and potential in the field. Expert Opin Investig Drugs. 16:1933–1953. 2007.
Article
33. Stack EC, Kubilus JK, Smith K, Cormier K, Signore SJD, Guelin E, et al. Chronology of behavioral symptoms and neuropathological sequela in R6/2 Huntington’s disease transgenic mice. J Comp Neurol. 490:354–370. 2005.
Article
34. Vorisek I, Syka M, Vargova L. Brain diffusivity and structural changes in the R6/2 mouse model of huntington disease. J Neurosci Res. 95:1474–1484. 2017.
Article
35. Wheeler VC, Auerbach W, White JK, Srinidhi J, Auerbach A, Ryan A, et al. Length-dependent gametic CAG repeat instability in the Huntington’s disease knock-in mouse. Hum Mol Genet. 8:115–122. 1999.
Article
36. White JK, Auerbach W, Duyao MP, Vonsattel JP, Gusella JF, Joyner AL, et al. Huntingtin is required for neurogenesis and is not impaired by the Huntington’s disease CAG expansion. Nat Genet. 17:404–410. 1997.
Article
Full Text Links
  • JKNS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr