We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

A novel gene therapy for neurodegenerative Lafora disease via EPM2A-loaded DLinDMA lipoplexes

    Hari Priya Vemana

    Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA

    ,
    Aishwarya Saraswat

    Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA

    ,
    Shraddha Bhutkar

    Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA

    ,
    Ketan Patel

    Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA

    &
    Vikas V Dukhande

    *Author for correspondence: Tel.: +1 7189902640;

    E-mail Address: dukhandv@stjohns.edu

    Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA

    Published Online:https://doi.org/10.2217/nnm-2020-0477

    Aim: To develop novel cationic liposomes as a nonviral gene delivery vector for the treatment of rare diseases, such as Lafora disease – a neurodegenerative epilepsy. Materials & methods: DLinDMA and DOTAP liposomes were formulated and characterized for the delivery of gene encoding laforin and expression of functional protein in HEK293 and neuroblastoma cells. Results: Liposomes with cationic lipids DLinDMA and DOTAP showed good physicochemical characteristics. Nanosized DLinDMA liposomes demonstrated desired transfection efficiency, negligible hemolysis and minimal cytotoxicity. Western blotting confirmed successful expression and glucan phosphatase assay demonstrated the biological activity of laforin. Conclusion: Our study is a novel preclinical effort in formulating cationic lipoplexes containing plasmid DNA for the therapy of rare genetic diseases such as Lafora disease.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Minassian BA. Lafora’s disease: towards a clinical, pathologic, and molecular synthesis. Pediatr. Neurol. 25(1), 21–29 (2001).
    • 2. Minassian BA, Lee JR, Herbrick JA et al. Mutations in a gene encoding a novel protein tyrosine phosphatase cause progressive myoclonus epilepsy. Nat. Genet. 20(2), 171–174 (1998). •• Demonstrates that mutations in EPM2A gene result in detrimental effects in laforin protein, which effects glycogen metabolism and causes progressive myoclonus epilepsy of Lafora type.
    • 3. Minassian BA, Ianzano L, Meloche M et al. Mutation spectrum and predicted function of laforin in Lafora’s progressive myoclonus epilepsy. Neurology 55(3), 341–346 (2000).
    • 4. Chan EM, Young EJ, Ianzano L et al. Mutations in NHLRC1 cause progressive myoclonus epilepsy. Nat. Genet. 35(2), 125–127 (2003).
    • 5. Worby CA, Gentry MS, Dixon JE. Laforin, a dual specificity phosphatase that dephosphorylates complex carbohydrates. J. Biol. Chem. 281(41), 30412–30418 (2006).
    • 6. Gentry MS, Worby CA, Dixon JE. Insights into Lafora disease: malin is an E3 ubiquitin ligase that ubiquitinates and promotes the degradation of laforin. Proc. Natl Acad. Sci. USA 102(24), 8501–8506 (2005).
    • 7. Wang W, Roach PJ. Glycogen and related polysaccharides inhibit the laforin dual-specificity protein phosphatase. Biochem. Biophys. Res. Commun. 325(3), 726–730 (2004).
    • 8. Gentry MS, Dowen RH 3rd, Worby CA, Mattoo S, Ecker JR, Dixon JE. The phosphatase laforin crosses evolutionary boundaries and links carbohydrate metabolism to neuronal disease. J. Cell Biol. 178(3), 477–488 (2007).
    • 9. Sullivan M, Nitschke S, Steup M, Minassian B, Nitschke F. Pathogenesis of Lafora disease: transition of soluble glycogen to insoluble polyglucosan. IJMS 18(8), 1743 (2017).
    • 10. Dukhande VV, Sherwood AR, Gentry MS. Laforin. UCSD-Nature Molecule Pages doi:10.1038/mp.a000032.01 (2010).
    • 11. Gentry MS, Romá-Mateo C, Sanz P. Laforin, a protein with many faces: glucan phosphatase, adapter protein, et alii. FEBS J. 280(2), 525–537 (2013). • Provides a review summary that laforin is involved in several physiological processes in addition to its phosphatase activity.
    • 12. Duran J, Gruart A, Garcia-Rocha M, Delgado-Garcia JM, Guinovart JJ. Glycogen accumulation underlies neurodegeneration and autophagy impairment in Lafora disease. Hum. Mol. Genet. 23(12), 3147–3156 (2014).
    • 13. DePaoli-Roach AA, Tagliabracci VS, Segvich DM, Meyer CM, Irimia JM, Roach PJ. Genetic depletion of the malin e3 ubiquitin ligase in mice leads to Lafora bodies and the accumulation of insoluble laforin. J. Biol. Chem. 285(33), 25372–25381 (2010).
    • 14. Raththagala M, Brewer MK, Parker MW et al. Structural mechanism of laforin function in glycogen dephosphorylation and Lafora disease. Mol. Cell 57(2), 261–272 (2015).
    • 15. Nitschke F, Wang P, Schmieder P et al. Hyperphosphorylation of glucosyl C6 carbons and altered structure of glycogen in the neurodegenerative epilepsy Lafora disease. Cell Metab. 17(5), 756–767 (2013).
    • 16. Nitschke F, Ahonen SJ, Nitschke S, Mitra S, Minassian BA. Lafora disease - from pathogenesis to treatment strategies. Nat. Rev. Neurol. 14(10), 606–617 (2018).
    • 17. Goldenberg MM. Overview of drugs used for epilepsy and seizures. P T. 35(7), 392–415 (2010).
    • 18. Michelucci R, Pasini E, Riguzzi P, Andermann E, Kälviäinen R, Genton P. Myoclonus and seizures in progressive myoclonus epilepsies: pharmacology and therapeutic trials. Epileptic Disord. 18(S2), 145–153 (2016).
    • 19. Goldsmith D, Minassian BA. Efficacy and tolerability of perampanel in ten patients with Lafora disease. Epilepsy Behav. 62, 132–135 (2016).
    • 20. Yang Y, Zhu B, Zheng F et al. Chronic metformin treatment facilitates seizure termination. Biochem. Biophys. Res. Commun. 484(2), 450–455 (2017).
    • 21. Mikati MA, Tabbara F. Managing Lafora body disease with vagal nerve stimulation. Epileptic Disord. 19(1), 82–86 (2017).
    • 22. Hajnsek S, Petelin Gadze Z, Borovecki F et al. Vagus nerve stimulation in Lafora body disease. Epilepsy Behav. Case Rep. 1, 150–152 (2013).
    • 23. Bennett CF, Krainer AR, Cleveland DW. Antisense oligonucleotide therapies for neurodegenerative diseases. Annu. Rev. Neurosci. 42(1), 385–406 (2019).
    • 24. Goldsmith D, Minassian BA. Extraneurological sparing in long-lived typical Lafora disease. Epilepsia Open 3(2), 295 (2018).
    • 25. Brewer MK, Uittenbogaard A, Austin GL et al. Targeting pathogenic Lafora bodies in Lafora disease using an antibody-enzyme fusion. Cell Metab. 30(4), 689–705.e6 (2019). •• Confirms that α-amylase is capable of degrading Lafora bodies and antibody enzyme fusion therapy can be used for the treatment of Lafora disease.
    • 26. Austin GL, Simmons ZR, Klier JE et al. Central nervous system delivery and biodistribution analysis of an antibody–enzyme fusion for the treatment of Lafora disease. Mol. Pharmaceutics 16(9), 3791–3801 (2019).
    • 27. Shahryari A, Saghaeian Jazi M, Mohammadi S et al. Development and clinical translation of approved gene therapy products for genetic disorders. Front. Genet. 10, 868 (2019).
    • 28. Bates M. Advances in gene therapy offer hope for rare disorders. IEEE Pulse 10(6), 9–12 (2019).
    • 29. Lechardeur D, Lukacs GL. Intracellular barriers to non-viral gene transfer. Curr. Gene Ther. 2(2), 183–194 (2002).
    • 30. Ruponen M, Honkakoski P, Rönkkö S, Pelkonen J, Tammi M, Urtti A. Extracellular and intracellular barriers in non-viral gene delivery. J. Control. Rel. 93(2), 213–217 (2003).
    • 31. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat. Rev. Genet. 4(5), 346–358 (2003).
    • 32. Ramamoorth M, Narvekar A. Non viral vectors in gene therapy - an overview. J. Clin. Diagn. Res. 9(1), GE01–GE06 (2015).
    • 33. Patil S, Gao Y-G, Lin X et al. The Development of functional non-viral vectors for gene delivery. Int. J. Mol. Sci. 20(21), 5491 (2019).
    • 34. Faheem AM, Elkordy AA, Hassan E, Carr-Wilkinson J. Preparation, characterisation and cell transfection of cationic liposomes in gene therapy. Br. J. Pharm. 4(1), 618 (2019).
    • 35. Shim G, Kim M-G, Park JY, Oh Y-K. Application of cationic liposomes for delivery of nucleic acids. Asian J. Pharm. Sci. 8(2), 72–80 (2013). • Demonstrates the importance and advantages of cationic liposomes for gene delivery, which could help in developing the treatment strategies for rare genetic disorders.
    • 36. Bae Y-U, Huh J-W, Kim B-K, Park HY, Seu Y-B, Doh K-O. Enhancement of liposome mediated gene transfer by adding cholesterol and cholesterol modulating drugs. Biochim. Biophys. Acta 1858(12), 3017–3023 (2016).
    • 37. Du Z, Munye MM, Tagalakis AD, Manunta MDI, Hart SL. The role of the helper lipid on the DNA transfection efficiency of lipopolyplex formulations. Sci. Rep. 4(1), 7107 (2014).
    • 38. Dukhande VV, Rogers DM, Roma-Mateo C et al. Laforin, a dual specificity phosphatase involved in Lafora disease, is present mainly as monomeric form with full phosphatase activity. PLoS ONE 6(8), e24040 (2011).
    • 39. Sherwood AR, Johnson MB, Delgado-Escueta AV, Gentry MS. A bioassay for Lafora disease and laforin glucan phosphatase activity. Clin. Biochem. 46(18), 1869–1876 (2013).
    • 40. Alton EWFW, Armstrong DK, Ashby D et al. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir. Med. 3(9), 684–691 (2015).
    • 41. McNamara EL, Taylor RL, Clayton JS et al. Systemic AAV8-mediated delivery of a functional copy of muscle glycogen phosphorylase (Pygm) ameliorates disease in a murine model of McArdle disease. Hum. Mol. Genet. 29(1), 20–30 (2020).
    • 42. Tang B, Frasinyuk MS, Chikwana VM et al. Discovery and development of small-molecule inhibitors of glycogen synthase. J. Med. Chem. 63(7), 3538–3551 (2020). • Proves the important role of glycogen synthase inhibitors in suppressing accumulation of glycogen, which could be a potential treatment of Lafora disease.
    • 43. Sánchez-Elexpuru G, Serratosa JM, Sanz P, Sánchez MP. 4-Phenylbutyric acid and metformin decrease sensitivity to pentylenetetrazol-induced seizures in a malin knockout model of Lafora disease. Neuroreport 28(5), 268–271 (2017).
    • 44. Cardinali S, Canafoglia L, Bertoli S et al. A pilot study of a ketogenic diet in patients with Lafora body disease. Epilepsy Res. 69(2), 129–134 (2006).
    • 45. Israelian L, Wang P, Gabrielian S, Zhao X, Minassian BA. Ketogenic diet reduces Lafora bodies in murine Lafora disease. Neurol. Genet. 6(6), e533 (2020).
    • 46. Hospital for Sick Children HSC. Ionis Pharmaceuticals Inc.: US20190194666 (2019).
    • 47. Rich LR, Harris W, Brown AM. The role of brain glycogen in supporting physiological function. Front. Neurosci. 13, 1176 (2019).
    • 48. Wolfsdorf JI, Weinstein DA. Glycogen storage diseases. Rev. Endocr. Metab. Disord. 4(1), 95–102 (2003).
    • 49. Colella P, Ronzitti G, Mingozzi F. Emerging issues in AAV-mediated in vivo gene therapy. Mol. Ther. Methods Clin. Dev. 8, 87–104 (2017).
    • 50. Ye T, Zhong Z, García-Sastre A, Schotsaert M, Geest BGD. Current status of COVID-19 (Pre)clinical vaccine development. Angew. Chem. Int. Ed. Engl. 59(43), 18885–18897 (2020).
    • 51. Brewer MK, Gentry MS. The 3rd International Lafora Workshop: evidence for a cure. Epilepsy Behav. 81, 125–127 (2018). •• Provides evidence that injection of antisense oligonucleotides targeting glycogen synthase reduced Lafora bodies and a natural history study on a small cohort of patients is initiated by Ionis Pharmaceuticals.