Therapeutic reversal of Huntington's disease by in vivo self-assembled siRNAs

Li Zhang, Tengteng Wu, Yangyang Shan, Ge Li, Xue Ni, Xiaorui Chen, Xiuting Hu, Lishan Lin, Yongchao Li, Yalun Guan, Jinfeng Gao, Dingbang Chen, Yu Zhang, Zhong Pei, Xi Chen, Li Zhang, Tengteng Wu, Yangyang Shan, Ge Li, Xue Ni, Xiaorui Chen, Xiuting Hu, Lishan Lin, Yongchao Li, Yalun Guan, Jinfeng Gao, Dingbang Chen, Yu Zhang, Zhong Pei, Xi Chen

Abstract

Huntington's disease is an autosomal-dominant neurodegenerative disease caused by CAG expansion in exon 1 of the huntingtin (HTT) gene. Since mutant huntingtin (mHTT) protein is the root cause of Huntington's disease, oligonucleotide-based therapeutic approaches using small interfering RNAs (siRNAs) and antisense oligonucleotides designed to specifically silence mHTT may be novel therapeutic strategies for Huntington's disease. Unfortunately, the lack of an effective in vivo delivery system remains a major obstacle to realizing the full potential of oligonucleotide therapeutics, especially regarding the delivery of oligonucleotides to the cortex and striatum, the most severely affected brain regions in Huntington's disease. In this study, we present a synthetic biology strategy that integrates the naturally existing exosome-circulating system with artificial genetic circuits for self-assembly and delivery of mHTT-silencing siRNA to the cortex and striatum. We designed a cytomegalovirus promoter-directed genetic circuit encoding both a neuron-targeting rabies virus glycoprotein tag and an mHTT siRNA. After being taken up by mouse livers after intravenous injection, this circuit was able to reprogramme hepatocytes to transcribe and self-assemble mHTT siRNA into rabies virus glycoprotein-tagged exosomes. The mHTT siRNA was further delivered through the exosome-circulating system and guided by a rabies virus glycoprotein tag to the cortex and striatum. Consequently, in three mouse models of Huntington's disease treated with this circuit, the levels of mHTT protein and toxic aggregates were successfully reduced in the cortex and striatum, therefore ameliorating behavioural deficits and striatal and cortical neuropathologies. Overall, our findings establish a convenient, effective and safe strategy for self-assembly of siRNAs in vivo that may provide a significant therapeutic benefit for Huntington's disease.

Keywords: Huntington’s disease; exosome; self-assembly; siRNA; synthetic biology.

© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.

Figures

Figure 1
Figure 1
Schematic description of the architecture of the genetic circuit. The genetic circuit contains three different functional modules: a co-driving CMV promoter, a neuron-guiding RVG tag and an mHTT siRNA-expressing backbone. When the genetic circuit is placed in a tissue chassis such as the liver, the CMV promoter drives the transcription of mHTT siRNA and facilitates loading of the payload of mHTT siRNA into exosomes as cargo; simultaneously, the CMV promoter directs the localization of the RVG guidance tag onto the exosome surface to confer neuron-targeting properties on the exosomes. After being released into the circulation and delivered through the exosome-circulating system, the mHTT siRNA enclosed in RVG-tagged exosomes is guided by the RVG tag to penetrate the blood–brain barrier (BBB) and arrive at the cortex and striatum, ultimately resulting in mRNA degradation and decreased protein expression of the mHTT gene.
Figure 2
Figure 2
Characterization of self-assembled mHTT siRNA in an ex vivo model. (A) Schematic of the experimental design. C57BL/6J mice were intravenously injected with PBS or with 5 mg/kg CMV-scrR, CMV-siRmHTT or CMV-RVG-siRmHTT circuit every 2 days for a total of seven times, and then the exosomes were purified from mouse plasma and incubated with HEK293T cells. Next, uptake of self-assembled mHTT siRNA by HEK293T cells and the subsequent suppression of mHTT expression and aggregation by self-assembled mHTT siRNA were examined in this ex vivo model. (B) Quantitative RT-PCR analysis of mHTT siRNA levels in purified exosomes (n =5 in each group). (C) Purified exosomes were fluorescently labelled with PKH26, and PKH26-labelled exosomes were incubated with HEK293T cells for 6 h. The levels of intracellular fluorescence intensity were monitored by confocal microscopy. (D) Quantitation of the fluorescence intensity shown in C (n =5 in each group). (E) Western blot analysis of mHTT-FLAG protein levels in mHTT-Q66-expressing HEK293T cells after incubation with purified exosomes. HEK293T cells were stably transfected with LV-mHTT-Q66-FLAG ahead of incubation. (F) Quantitation of the mHTT-FLAG protein levels shown in E (n =5 in each group). (G) Quantitative RT-PCR analysis of mHTT mRNA levels in mHTT-Q66-expressing HEK293T cells after incubation with purified exosomes (n =5 in each group). (H) HEK293T cells coexpressing mHTT-Q66 and GFP were incubated with purified exosomes. Fluorescence microscopy was used to assess mHTT aggregates (bright green puncta). Scale bar = 5 μm. (I) Quantitation of the aggregated mHTT dots shown in H (n =5 in each group). (J) Purified exosomes were fluorescently labelled with PKH26, and PKH26-labelled exosomes were incubated with STHdh-Q111 cells for 6 h. The levels of intracellular fluorescence intensity were monitored by confocal microscopy. Scale bar = 20 μm. (K) Western blot analysis of mHTT protein levels in STHdh-Q111 cells after incubation with purified exosomes. STHdh-Q7 cells expressing wild-type HTT served as a control. (L) Quantitation of the HTT protein levels shown in K (n =5 in each group). Values are presented as mean ± SEM. Significance was determined using one-way ANOVA followed by Bonferroni's multiple comparisons in F, G, I and L. **P <0.01; ***P <0.001; ****P <0.0001; ns = not significant.
Figure 3
Figure 3
Tracking and visualization of the delivery of self-assembled mHTT siRNA into the cortex and striatum. (AC) Quantitative RT–PCR analysis of mHTT siRNA levels in the liver, plasma and cortex/striatum after intravenous injection of C57BL/6J mice with PBS or with 5 mg/kg CMV-scrR, CMV-siRmHTT or CMV-RVG-siRmHTT circuit for a total of three times (n =5 in each group). Values are presented as the means ± SEM. (D and E) In situ detection of mHTT siRNA in the mouse cortex and striatum. C57BL/6J mice were intravenously injected with PBS or with 5 mg/kg CMV-scrR, CMV-siRmHTT or CMV-RVG-siRmHTT circuit for a total of three times. Positive in situ hybridization signals in the cortex and striatum are shown in green, and DAPI-stained nuclei are shown in blue. Scale bar = 50 μm. (F and G) Direct visualization of the suppression of GFP fluorescence levels in vivo by self-assembled GFP siRNA. GFP-transgenic mice were intravenously injected with PBS or with 5 mg/kg CMV-scrR, CMV-siRGFP or CMV-RVG-siRGFP circuit every 2 days for a total of seven times. After treatment termination, mice were killed and GFP fluorescence levels were assessed in frozen sections of the cortex and striatum. Representative fluorescence microscopy images are shown. Positive GFP signals are shown in green, NeuN-stained neurons are shown in red and DAPI-stained nuclei are shown in blue. Scale bar = 100 μm.
Figure 4
Figure 4
Evaluation of the therapeutic efficacy of self-assembled mHTT siRNA in N171-82Q and BACHD Huntington’s disease models. (A) Schematic of the experimental design. At 8 weeks of age, N171-82Q mice were intravenously injected with 5 mg/kg CMV-scrR or CMV-RVG-siRmHTT circuit every 2 days for a total of 2 weeks. After treatment termination, the mice were monitored to evaluate behavioural performance and mHTT accumulation. (B) Motor performance in a rotarod test. Non-transgenic littermates were included as behavioural controls (n =8 in each group). (C) Western blot analysis of mHTT protein levels in the cortex and striatum. α-Tubulin served as the internal loading control. (D) Quantitation of the mHTT protein levels shown in C (n =4 in cortex; n =8 in striatum). (E) Quantitative RT-PCR analysis of mHTT mRNA levels in the cortex and striatum (n =7–8 in each group). (F) Schematic of the experimental design. At 3 months of age, BACHD mice were intravenously injected with 5 mg/kg CMV-scrR or CMV-RVG-siRmHTT circuit every 2 days for a total of 2 weeks. After treatment termination, the mice were monitored to evaluate mHTT accumulation and neuropathology. (G) Western blot analysis of mHTT protein levels in the cortex and striatum. The more slowly migrating band is the human mHTT protein, and the more rapidly migrating band is mouse endogenous HTT. Endogenous HTT and α-tubulin served as the internal loading control. (H) Quantitation of the mHTT protein levels shown in G (n =8 in each group). (I) Quantitative RT–PCR analysis of mHTT mRNA levels in the cortex and striatum (n =8 in each group). (J) Immunofluorescence staining of aggregated mHTT (EM48, green), neurons (NeuN, red) and nuclei (DAPI, blue) in striatal and cortical sections. Scale bar = 50 μm. Values are presented as the means ± SEM. Significance was determined using two-sided t-test in D, E, H and I, or using one-way ANOVA followed by Bonferroni's multiple comparisons in B. *P <0.05; ***P <0.001; ****P <0.0001; ns = not significant.
Figure 5
Figure 5
Evaluation of the long-term therapeutic efficacy of self-assembled mHTT siRNA in the YAC128 Huntington’s disease model. (A) Schematic of the experimental design. At 6 weeks of age, YAC128 mice were intravenously injected with 5 mg/kg CMV-scrR or CMV-RVG-siRmHTT circuit twice a week for a total of 8 weeks. After treatment termination, the mice were monitored to evaluate behavioural performance, mHTT accumulation and neuropathology. (B) Motor performance on a rotarod test. Non-transgenic littermates were included as behavioural controls (n =8 in each group). (C) Western blot analysis of mHTT protein levels in the cortex and striatum. The more slowly migrating band is the human mHTT protein, and the more rapidly migrating band is mouse endogenous HTT. Endogenous HTT served as the internal loading control. (D) Quantitation of the mHTT protein levels shown in (C) (n =8 in each group). (E) Quantitative RT–PCR analysis of mHTT mRNA levels in the cortex and striatum (n =8 in each group). (F) Immunofluorescence staining of aggregated mHTT (EM48, green), neurons (NeuN, red) and nuclei (DAPI, blue) in striatal and cortical sections. Scale bar = 50 μm. Values are presented as mean ± SEM. Significance was determined using two-sided t-test in D and E, or using one-way ANOVA followed by Bonferroni's multiple comparisons in B. ***P <0.001; ****P <0.0001; ns = not significant.

References

    1. Novak MJ, Tabrizi SJ.. Huntington's disease. BMJ. 2010;340:c3109.
    1. Ross C, Aylward E, Wild E, et al.Huntington disease: Natural history, biomarkers and prospects for therapeutics. Nat Rev Neurol. 2014;10(4):204–216.
    1. Tabrizi S, Flower M, Ross C, Wild E.. Huntington disease: New insights into molecular pathogenesis and therapeutic opportunities. Nat Rev Neurol. 2020;16(10):529–546.
    1. MacDonald ME, Duyao MP, Myers RH, 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. 1993;72(6):971–983.
    1. Sathasivam K, Neueder A, Gipson T, et al.Aberrant splicing of HTT generates the pathogenic exon 1 protein in Huntington disease. Proc Natl Acad Sci U S A. 2013;110(6):2366–2370.
    1. Ross CA, Tabrizi SJ.. Huntington's disease: From molecular pathogenesis to clinical treatment. Lancet Neurol. 2011;10(1):83–98.
    1. Bates G. Huntingtin aggregation and toxicity in Huntington's disease. Lancet. 2003;361(9369):1642–1644.
    1. Walker FO. Huntington's disease. Lancet. 2007;369(9557):218–228.
    1. McColgan P, Tabrizi SJ.. Huntington's disease: A clinical review. Eur J Neurol. 2018;25(1):24–34.
    1. Sah D, Aronin N.. Oligonucleotide therapeutic approaches for Huntington disease. J Clin Invest. 2011;121(2):500–507.
    1. Davidson B, McCray P.. Current prospects for RNA interference-based therapies. Nat Rev Genet. 2011;12(5):329–340.
    1. Kole R, Krainer A, Altman S.. RNA therapeutics: Beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov. 2012;11(2):125–140.
    1. Rinaldi C, Wood M.. Antisense oligonucleotides: The next frontier for treatment of neurological disorders. Nat Rev Neurol. 2018;14(1):9–21.
    1. Xia H, Mao Q, Paulson HL, Davidson BL.. siRNA-mediated gene silencing in vitro and in vivo. Nat Biotechnol. 2002;20(10):1006–1010.
    1. Yu D, Pendergraff H, Liu J, et al.Single-stranded RNAs use RNAi to potently and allele-selectively inhibit mutant huntingtin expression. Cell. 2012;150(5):895–908.
    1. Kordasiewicz H, Stanek L, Wancewicz E, et al.Sustained therapeutic reversal of Huntington's disease by transient repression of huntingtin synthesis. Neuron. 2012;74(6):1031–1044.
    1. Svrzikapa N, Longo K, Prasad N, et al.Investigational assay for haplotype phasing of the huntingtin gene. Mol Ther Methods Clin Dev. 2020;19:162–173.
    1. Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, et al.Targeting huntingtin expression in patients with Huntington's disease. N Engl J Med. 2019;380(24):2307–2316.
    1. Gomes M, Martins S, Sarmento B.. siRNA as a tool to improve the treatment of brain diseases: Mechanism, targets and delivery. Ageing Res Rev. 2015;21:43–54.
    1. Pardridge WM. shRNA and siRNA delivery to the brain. Adv Drug Deliv Rev. 2007;59(2-3):141–152.
    1. Caron NS, Dorsey ER, Hayden MR.. Therapeutic approaches to Huntington disease: From the bench to the clinic. Nat Rev Drug Discov. 2018;17(10):729–750.
    1. Roberts T, Langer R, Wood M.. Advances in oligonucleotide drug delivery. Nat Rev Drug Discov. 2020;19(10):673–694.
    1. Kalluri R, LeBleu V.. The biology function and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977
    1. O'Brien K, Breyne K, Ughetto S, Laurent L, Breakefield X.. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 2020;21(10):585–606.
    1. Zhang Y, Liu D, Chen X, et al.Secreted monocytic miR-150 enhances targeted endothelial cell migration. Mol Cell. 2010;39(1):133–144.
    1. Li J, Zhang Y, Liu Y, et al.Microvesicle-mediated transfer of microRNA-150 from monocytes to endothelial cells promotes angiogenesis. J Biol Chem. 2013;288(32):23586–23596.
    1. Yin Y, Cai X, Chen X, et al.Tumor-secreted miR-214 induces regulatory T cells: A major link between immune evasion and tumor growth. Cell Res. 2014;24(10):1164–1180.
    1. Zhou Y, Zhou G, Tian C, et al.Exosome-mediated small RNA delivery for gene therapy. Wiley Interdiscip Rev RNA. 2016;7(6):758–771.
    1. van Niel G, D'Angelo G, Raposo G.. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–228.
    1. Syn N, Wang L, Chow E, Lim C, Goh B.. Exosomes in cancer nanomedicine and immunotherapy: prospects and challenges. Trends Biotechnol. 2017;35(7):665–676.
    1. Kitada T, DiAndreth B, Teague B, Weiss R.. Programming gene and engineered-cell therapies with synthetic biology. Science. 2018;359(6376):eaad1067.
    1. Weber W, Fussenegger M.. Emerging biomedical applications of synthetic biology. Nat Rev Genet. 2011;13(1):21–35.
    1. Atsumi S, Hanai T, Liao J.. Non-fermentative pathways for synthesis of branched-chain higher alcohols as biofuels. Nature. 2008;451(7174):86–89.
    1. Elani Y. Interfacing living and synthetic cells as an emerging frontier in synthetic biology. Angew Chem Int Ed Engl. 2021;60(11):5602–5611.
    1. Fu Z, Zhang X, Zhou X, et vivo self-assembled small RNAs as a new generation of RNAi therapeutics. Cell Res. 2021;31(6):631–648.
    1. Liu F, Song Y, Liu D.. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Ther. 1999;6(7):1258–1266.
    1. Zhang G, Budker V, Wolff J.. High levels of foreign gene expression in hepatocytes after tail vein injections of naked plasmid DNA. Hum Gene Therapy. 1999;10(10):1735–1737.
    1. Tiago Ferreira WR. ImageJ User Guide.
    1. Marco S, Giralt A, Petrovic M, et al.Suppressing aberrant GluN3A expression rescues synaptic and behavioral impairments in Huntington's disease models. Nat Med. 2013;19(8):1030–1038.
    1. Fox L, Kim K, Johnson C, et al.Huntington's disease pathogenesis is modified in vivo by Alfy/Wdfy3 and selective macroautophagy. Neuron. 2020;105(5):813–821.e6.
    1. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood M.. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–345.
    1. DiFiglia M, Sena-Esteves M, Chase K, et al.Therapeutic silencing of mutant huntingtin with siRNA attenuates striatal and cortical neuropathology and behavioral deficits. Proc Natl Acad Sci U S A. 2007;104(43):17204–17209.
    1. Hodgson J, Agopyan N, Gutekunst C, et al.A YAC mouse model for Huntington's disease with full-length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurodegeneration. Neuron. 1999;23(1):181–192.
    1. Menalled LB, Chesselet M-F.. Mouse models of Huntington's disease. Trends Pharmacol Sci. 2002;23(1):32–39.
    1. Beal MF, Ferrante RJ.. Experimental therapeutics in transgenic mouse models of Huntington's disease. Nat Rev Neurosci. 2004;5(5):373–384.
    1. Wang N, Gray M, Lu X, et al.Neuronal targets for reducing mutant huntingtin expression to ameliorate disease in a mouse model of Huntington's disease. Nat Med. 2014;20(5):536–541.
    1. Gu X, Cantle J, Greiner E, et al.N17 Modifies mutant huntingtin nuclear pathogenesis and severity of disease in HD BAC transgenic mice. Neuron. 2015;85(4):726–741.
    1. Pouladi MA, Morton AJ, Hayden MR.. Choosing an animal model for the study of Huntington's disease. Nat Rev Neurosci. 2013;14(10):708–721.
    1. Kim B, Park J, Sailor M.. Rekindling RNAi therapy: Materials design requirements for in vivo siRNA delivery. Adv Mater. 2019;31(49):e1903637.
    1. McKenzie LK, El-Khoury R, Thorpe JD, Damha MJ, Hollenstein M.. Recent progress in non-native nucleic acid modifications. Chem Soc Rev. 2021;50(8):5126–5164.
    1. Vogel G. Gene therapy. FDA moves against Penn scientist. Science. 2000;290(5499):2049–2051.
    1. Zuckerman J, Gritli I, Tolcher A, et al.Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc Natl Acad Sci U S A. 2014;111(31):11449–11454.
    1. Gabisonia K, Prosdocimo G, Aquaro G, et al.MicroRNA therapy stimulates uncontrolled cardiac repair after myocardial infarction in pigs. Nature. 2019;569(7756):418–422.
    1. Wu T, Su F, Feng Y, et al.Mesenchymal stem cells alleviate AQP-4-dependent glymphatic dysfunction and improve brain distribution of antisense oligonucleotides in BACHD mice. Stem Cells. 2020;38(2):218–230.
    1. Khalil A, Collins J.. Synthetic biology: Applications come of age. Nat Rev Genet. 2010;11(5):367–379.
    1. Tan X, Letendre J, Collins J, Wong W.. Synthetic biology in the clinic: Engineering vaccines, diagnostics, and therapeutics. Cell. 2021;184(4):881–898.
    1. Glass D, Alon U.. Programming cells and tissues. Science. 2018;361(6408):1199–1200.
    1. Lentz T, Wilson P, Hawrot E, Speicher D.. Amino acid sequence similarity between rabies virus glycoprotein and snake venom curaremimetic neurotoxins. Science. 1984;226(4676):847–848.
    1. Kumar P, Wu H, McBride J, et al.Transvascular delivery of small interfering RNA to the central nervous system. Nature. 2007;448(7149):39–43.
    1. Lentz T, Burrage T, Smith A, Crick J, Tignor G.. Is the acetylcholine receptor a rabies virus receptor? Science. 1982;215(4529):182–184.
    1. Mulcahy LA, Pink RC, Carter DRF.. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014. Aug 4;3:10.3402/jev.v3.24641.
    1. El Andaloussi S, Lakhal S, Mäger I, Wood MJA.. Exosomes for targeted siRNA delivery across biological barriers. Adv Drug Deliv Rev. 2013;65(3):391–397.
    1. Morad G, Carman CV, Hagedorn EJ, et al.Tumor-derived extracellular vesicles breach the intact blood-brain barrier transcytosis. ACS Nano. 2019;13(12):13853–13865.
    1. Rodrigues G, Hoshino A, Kenific CM, et al.Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis. Nat Cell Biol. 2019;21(11):1403–1412.
    1. Men Y, Yelick J, Jin S, et al.Exosome reporter mice reveal the involvement of exosomes in mediating neuron to astroglia communication in the CNS. Nat Commun. 2019;10(1):4136.

Source: PubMed

3
Abonnere