
- Select a language for the TTS:
- UK English Female
- UK English Male
- US English Female
- US English Male
- Australian Female
- Australian Male
- Language selected: (auto detect) - EN
Play all audios:
ABSTRACT Sustained silencing of gene expression throughout the brain using small interfering RNAs (siRNAs) has not been achieved. Here we describe an siRNA architecture, divalent siRNA
(di-siRNA), that supports potent, sustained gene silencing in the central nervous system (CNS) of mice and nonhuman primates following a single injection into the cerebrospinal fluid.
Di-siRNAs are composed of two fully chemically modified, phosphorothioate-containing siRNAs connected by a linker. In mice, di-siRNAs induced the potent silencing of huntingtin, the
causative gene in Huntington’s disease, reducing messenger RNA and protein throughout the brain. Silencing persisted for at least 6 months, with the degree of gene silencing correlating to
levels of guide strand tissue accumulation. In cynomolgus macaques, a bolus injection of di-siRNA showed substantial distribution and robust silencing throughout the brain and spinal cord
without detectable toxicity and with minimal off-target effects. This siRNA design may enable RNA interference-based gene silencing in the CNS for the treatment of neurological disorders.
Access through your institution Buy or subscribe This is a preview of subscription content, access via your institution ACCESS OPTIONS Access through your institution Access Nature and 54
other Nature Portfolio journals Get Nature+, our best-value online-access subscription $29.99 / 30 days cancel any time Learn more Subscribe to this journal Receive 12 print issues and
online access $209.00 per year only $17.42 per issue Learn more Buy this article * Purchase on SpringerLink * Instant access to full article PDF Buy now Prices may be subject to local taxes
which are calculated during checkout ADDITIONAL ACCESS OPTIONS: * Log in * Learn about institutional subscriptions * Read our FAQs * Contact customer support SIMILAR CONTENT BEING VIEWED BY
OTHERS CHEMICAL ENGINEERING OF THERAPEUTIC SIRNAS FOR ALLELE-SPECIFIC GENE SILENCING IN HUNTINGTON’S DISEASE MODELS Article Open access 03 October 2022 DEVELOPMENT OF AN AAV-RNAI STRATEGY TO
SILENCE THE DOMINANT VARIANT _GNAO1_ C.607G>A LINKED TO ENCEPHALOPATHY Article 14 April 2025 THE THERAPEUTIC IMPLICATIONS OF _ALL-IN-ONE_ AAV-DELIVERED EPIGENOME-EDITING PLATFORM IN
NEURODEGENERATIVE DISORDERS Article Open access 23 August 2024 DATA AVAILABILITY The RNA-seq data from nonhuman primate samples have been deposited in GEO under accession code GSE130132,
including processed transcriptome read counts. REFERENCES * Khvorova, A. & Watts, J. K. The chemical evolution of oligonucleotide therapies of clinical utility. _Nat. Biotechnol._ 35,
238–248 (2017). Article CAS Google Scholar * Ostergaard, M. E. et al. Efficient synthesis and biological evaluation of 5’-galnac conjugated antisense oligonucleotides. _Bioconjug. Chem._
26, 1451–1455 (2015). Article Google Scholar * Rajeev, K. G. et al. Hepatocyte-specific delivery of siRNAs conjugated to novel non-nucleosidic trivalent N-acetylgalactosamine elicits
robust gene silencing in vivo. _Chembiochem._ 16, 903–908 (2015). Article CAS Google Scholar * Kordasiewicz, H. B. et al. Sustained therapeutic reversal of Huntington’s disease by
transient repression of huntingtin synthesis. _Neuron._ 74, 1031–1044 (2012). Article CAS Google Scholar * Finkel, R. S. et al. Nusinersen versus sham control in infantile-onset spinal
muscular atrophy. _N. Engl. J. Med._ 377, 1723–1732 (2017). Article CAS Google Scholar * Coelho, T. et al. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. _N. Engl. J.
Med._ 369, 819–829 (2013). Article CAS Google Scholar * Alterman, J. F. et al. Hydrophobically modified siRNAs silence huntingtin mRNA in primary neurons and mouse brain. _Mol. Ther.
Nucleic Acids_ 4, e266 (2015). Article CAS Google Scholar * Nikan, M. et al. Docosahexaenoic acid conjugation enhances distribution and safety of siRNA upon local administration in mouse
brain. _Mol. Ther. Nucleic Acids_ 5, e344 (2016). Article CAS Google Scholar * Osborn, M. F. & Khvorova, A. Improving small interfering RNA delivery in vivo through lipid conjugation.
_Nucleic Acid Ther_. 28, 128–136 (2018). Article CAS Google Scholar * Nikan, M. et al. Synthesis and evaluation of parenchymal retention and efficacy of a metabolically stable
O-Phosphocholine-N-docosahexaenoyl-l-serine siRNA conjugate in mouse brain. _Bioconjug. Chem._ 28, 1758–1766 (2017). Article CAS Google Scholar * Ly, S. et al. Visualization of
self-delivering hydrophobically modified siRNA cellular internalization. _Nucleic Acids Res._ 45, 15–25 (2017). Article CAS Google Scholar * Eckstein, F. Phosphorothioates, essential
components of therapeutic oligonucleotides. _Nucleic Acid Ther._ 24, 374–387 (2014). Article CAS Google Scholar * Flierl, U. et al. Phosphorothioate backbone modifications of
nucleotide-based drugs are potent platelet activators. _J. Exp. Med._ 212, 129–137 (2015). Article CAS Google Scholar * Sewing, S. et al. Assessing single-stranded oligonucleotide
drug-induced effects in vitro reveals key risk factors for thrombocytopenia. _PLoS ONE_ 12, e0187574 (2017). Article Google Scholar * Crooke, S. T., Wang, S., Vickers, T. A., Shen, W.
& Liang, X. H. Cellular uptake and trafficking of antisense oligonucleotides. _Nat. Biotechnol._ 35, 230–237 (2017). Article CAS Google Scholar * Hassler, M. R. et al. Comparison of
partially and fully chemically-modified siRNA in conjugate-mediated delivery in vivo. _Nucleic Acids Res._ 46, 2185–2196 (2018). Article CAS Google Scholar * Behlke, M. A. Progress
towards in vivo use of siRNAs. _Mol. Ther._ 13, 644–670 (2006). Article CAS Google Scholar * Winkler, J., Stessl, M., Amartey, J. & Noe, C. R. Off-target effects related to the
phosphorothioate modification of nucleic acids. _Chem. Med. Chem._ 5, 1344–1352 (2010). Article CAS Google Scholar * Amarzguioui, M., Holen, T., Babaie, E. & Prydz, H. Tolerance for
mutations and chemical modifications in a siRNA. _Nucl. Acids Res._ 31, 589–595 (2003). Article CAS Google Scholar * Harborth, J. et al. Sequence, chemical, and structural variation of
small interfering RNAs and short hairpin RNAs and the effect on mammalian gene silencing. _Antisense Nucl. Acid Drug Devel._ 13, 83–105 (2003). Article CAS Google Scholar * Nair, J. K. et
al. Impact of enhanced metabolic stability on pharmacokinetics and pharmacodynamics of GalNAc-siRNA conjugates. _Nucleic Acids Res._ 45, 10969–10977 (2017). Article CAS Google Scholar *
Fitzgerald, K. et al. A highly durable RNAi therapeutic inhibitor of PCSK9. _N. Engl. J. Med._ 376, 41–51 (2017). Article CAS Google Scholar * Ray, K. K. et al. Inclisiran in patients at
high cardiovascular risk with elevated LDL cholesterol. _N. Engl. J. Med._ 376, 1430–1440 (2017). Article CAS Google Scholar * Lee, Y. C. et al. Binding of synthetic oligosaccharides to
the hepatic Gal/GalNAc lectin. Dependence on fine structural features. _J. Biol. Chem._ 258, 199–202 (1983). CAS PubMed Google Scholar * Roehl, I., Schuster, M. & Seiffert, S. US
Patent 20110201006 A1 (2011). * Liu, C. C., Liu, C. C., Kanekiyo, T., Xu, H. & Bu, G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. _Nat. Rev. Neurol._ 9, 106–118
(2013). Article CAS Google Scholar * Mahley, R. W., Weisgraber, K. H. & Huang, Y. Apolipoprotein E4: a causative factor and therapeutic target in neuropathology, including
Alzheimer’s disease. _Proc. Natl Acad. Sci. USA_ 103, 5644–5651 (2006). Article CAS Google Scholar * Zetterberg, H., Jacobsson, J., Rosengren, L., Blennow, K. & Andersen, P. M.
Association of APOE with age at onset of sporadic amyotrophic lateral sclerosis. _J. Neurol. Sci._ 273, 67–69 (2008). Article CAS Google Scholar * Didiot, M. C. et al. Nuclear
localization of huntingtin mRNA Is specific to cells of neuronal origin. _Cell. Rep._ 24, 2553–2560 e2555 (2018). Article CAS Google Scholar * Achuta, V. S. et al. Tissue plasminogen
activator contributes to alterations of neuronal migration and activity-dependent responses in fragile X mice. _J. Neurosci._ 34, 1916–1923 (2014). Article CAS Google Scholar * Gu, X. et
al. N17 Modifies mutant huntingtin nuclear pathogenesis and severity of disease in HD BAC transgenic mice. _Neuron_ 85, 726–741 (2015). Article CAS Google Scholar * DiFiglia, M. et al.
Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain neurons. _Neuron_ 14, 1075–1081 (1995). Article CAS Google Scholar * Herndon, E. S. et al.
Neuroanatomic profile of polyglutamine immunoreactivity in Huntington disease brains. _J. Neuropathol. Exp. Neurol._ 68, 250–261 (2009). Article CAS Google Scholar * Weyer, A. &
Schilling, K. Developmental and cell type-specific expression of the neuronal marker NeuN in the murine cerebellum. _J. Neurosci. Res._ 73, 400–409 (2003). Article CAS Google Scholar *
Takala, R. S. et al. Glial fibrillary acidic protein and ubiquitin C-Terminal hydrolase-L1 as outcome predictors in traumatic brain injury. _World Neurosurg._ 87, 8–20 (2016). Article
Google Scholar * Lind, D., Franken, S., Kappler, J., Jankowski, J. & Schilling, K. Characterization of the neuronal marker NeuN as a multiply phosphorylated antigen with discrete
subcellular localization. _J. Neurosci. Res._ 79, 295–302 (2005). Article CAS Google Scholar * Dou, C. L., Li, S. & Lai, E. Dual role of brain factor-1 in regulating growth and
patterning of the cerebral hemispheres. _Cereb. Cortex_ 9, 543–550 (1999). Article CAS Google Scholar * Janas, M. M. et al. Selection of GalNAc-conjugated siRNAs with limited
off-target-driven rat hepatotoxicity. _Nat. Commun._ 9, 723 (2018). Article Google Scholar * Birmingham, A. et al. 3’ UTR seed matches, but not overall identity, are associated with RNAi
off-targets. _Nat. Methods_ 3, 199–204 (2006). Article CAS Google Scholar * Zuccato, C., Valenza, M. & Cattaneo, E. Molecular mechanisms and potential therapeutical targets in
Huntington’s disease. _Physiol. Rev._ 90, 905–981 (2010). Article CAS Google Scholar * Evers, M. M. et al. AAV5-miHTT gene therapy demonstrates broad distribution and strong human mutant
huntingtin lowering in a huntington’s disease minipig model. _Mol. Ther._ 26, 2163–2177 (2018). Article CAS Google Scholar * Southwell, A. L. et al. Huntingtin suppression restores
cognitive function in a mouse model of Huntington’s disease. _Sci. Transl. Med._ 10, eaar3959 (2018). Article Google Scholar * Pfister, E. L. et al. Artificial miRNAs reduce human mutant
huntingtin throughout the striatum in a transgenic sheep model of Huntington’s disease. _Hum. Gene. Ther._ 29, 663–673 (2018). Article CAS Google Scholar * Grondin, R. et al. Six-month
partial suppression of huntingtin is well tolerated in the adult rhesus striatum. _Brain_ 135, 1197–1209 (2012). Article Google Scholar * Tabrizi, S. et al. Effects of IONIS-HTTRx in
patients with early Huntington’s disease, results of the first HTT-lowering drug trial (CT.002). _Neurology_ 90, CT.002 (2018). Google Scholar * Bhagat, L. et al. Novel oligonucleotides
containing two 3’-ends complementary to target mRNA show optimal gene-silencing activity. _J. Med. Chem._ 54, 3027–3036 (2011). Article CAS Google Scholar * Malcolm, D. W., Sorrells, J.
E., Van Twisk, D., Thakar, J. & Benoit, D. S. Evaluating side effects of nanoparticle-mediated siRNA delivery to mesenchymal stem cells using next generation sequencing and enrichment
analysis. _Bioeng. Transl. Med._ 1, 193–206 (2016). Article CAS Google Scholar * Wang, G., Liu, X., Gaertig, M. A., Li, S. & Li, X. J. Ablation of huntingtin in adult neurons is
nondeleterious but its depletion in young mice causes acute pancreatitis. _Proc. Natl Acad. Sci. USA_ 113, 3359–3364 (2016). Article CAS Google Scholar * Pfister, E. L. et al. Five siRNAs
targeting three SNPs may provide therapy for three-quarters of Huntington’s disease patients. _Curr. Biol._ 19, 774–778 (2009). Article CAS Google Scholar * Godinho, B. et al.
Transvascular delivery of hydrophobically modified siRNAs: Gene silencing in the rat brain upon disruption of the blood-brain barrier. _Mol. Ther._ 26, 2580–2591 (2018). Article CAS Google
Scholar Download references ACKNOWLEDGEMENTS This project was funded by the NIH/NINDS (grant no. R01 NS104022; for A.K.), NIH/OD (grant no. S10 OD020012; for A.K.), CHDI (Research
Agreement no. A-6119; for N.A.), Alzheimer’s Drug Discovery Foundation (grant no. 20170101; for A.K.), Milton-Safenowitz Fellowship (no. 17-PDF-363; for B.M.D.C.G.) and The Berman–Topper
Fund (for A.K. and N.A.). We would like to thank the University of Massachusetts Medical School Animal Medicine Department and veterinary technicians for their contributions to the
large-animal studies. We would like to thank M.-C. Didiot for her mouse brain cartoon, E. Mohn and S. Hildebrand for editing the manuscript and Charles River Laboratories for help with
neuropathology. AUTHOR INFORMATION Author notes * These authors contributed equally: Julia F. Alterman, Bruno M.D.C. Godinho, Matthew R. Hassler, Chantal M. Ferguson. AUTHORS AND
AFFILIATIONS * RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA Julia F. Alterman, Bruno M. D. C. Godinho, Matthew R. Hassler, Chantal M. Ferguson,
Dimas Echeverria, Reka A. Haraszti, Andrew H. Coles, Faith Conroy, Rachael Miller, Loic Roux, Paul Yan, Emily G. Knox, Anton A. Turanov, Athma A. Pai, Neil Aronin & Anastasia Khvorova *
Department of Neurology, Massachusetts General Institute for Neurodegenerative Disease, Boston, MA, USA Ellen Sapp & Marian DiFiglia * Department of Medicine, University of Massachusetts
Medical School, Worcester, MA, USA Faith Conroy, Rachael Miller & Neil Aronin * Department of Radiology, New England Center for Stroke Research, University of Massachusetts Medical
School, Worcester, MA, USA Robert M. King, Heather L. Gray-Edwards & Matthew J. Gounis * Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA Robert
M. King * Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA Gwladys Gernoux, Christian Mueller & Miguel Sena-Esteves * Department of Pediatrics,
University of Massachusetts Medical School, Worcester, MA, USA Christian Mueller * Department of Neurosurgery, University of Massachusetts Medical School, Worcester, MA, USA Richard P. Moser
* Department of Animal Medicine, University of Massachusetts Medical School, Worcester, MA, USA Nina C. Bishop & Samer M. Jaber * Department of Pathology, University of Massachusetts
Medical School, Worcester, MA, USA Samer M. Jaber * Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA Miguel Sena-Esteves * Program in Molecular
Medicine, University of Massachusetts Medical School, Worcester, MA, USA Anastasia Khvorova Authors * Julia F. Alterman View author publications You can also search for this author inPubMed
Google Scholar * Bruno M. D. C. Godinho View author publications You can also search for this author inPubMed Google Scholar * Matthew R. Hassler View author publications You can also search
for this author inPubMed Google Scholar * Chantal M. Ferguson View author publications You can also search for this author inPubMed Google Scholar * Dimas Echeverria View author
publications You can also search for this author inPubMed Google Scholar * Ellen Sapp View author publications You can also search for this author inPubMed Google Scholar * Reka A. Haraszti
View author publications You can also search for this author inPubMed Google Scholar * Andrew H. Coles View author publications You can also search for this author inPubMed Google Scholar *
Faith Conroy View author publications You can also search for this author inPubMed Google Scholar * Rachael Miller View author publications You can also search for this author inPubMed
Google Scholar * Loic Roux View author publications You can also search for this author inPubMed Google Scholar * Paul Yan View author publications You can also search for this author
inPubMed Google Scholar * Emily G. Knox View author publications You can also search for this author inPubMed Google Scholar * Anton A. Turanov View author publications You can also search
for this author inPubMed Google Scholar * Robert M. King View author publications You can also search for this author inPubMed Google Scholar * Gwladys Gernoux View author publications You
can also search for this author inPubMed Google Scholar * Christian Mueller View author publications You can also search for this author inPubMed Google Scholar * Heather L. Gray-Edwards
View author publications You can also search for this author inPubMed Google Scholar * Richard P. Moser View author publications You can also search for this author inPubMed Google Scholar *
Nina C. Bishop View author publications You can also search for this author inPubMed Google Scholar * Samer M. Jaber View author publications You can also search for this author inPubMed
Google Scholar * Matthew J. Gounis View author publications You can also search for this author inPubMed Google Scholar * Miguel Sena-Esteves View author publications You can also search for
this author inPubMed Google Scholar * Athma A. Pai View author publications You can also search for this author inPubMed Google Scholar * Marian DiFiglia View author publications You can
also search for this author inPubMed Google Scholar * Neil Aronin View author publications You can also search for this author inPubMed Google Scholar * Anastasia Khvorova View author
publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS J.F.A., B.M.D.C.G., M.R.H. and A.K. conceived the project. J.F.A., B.M.D.C.G., M.R.H., C.M.F., M.D,
N.A. and A.K. contributed to the experimental design. J.F.A., B.M.D.C.G., C.M.F, E.S., C.M.F., R.A.H., A.H.C., F.C., R.M., L.R., P.Y., A.A.T., E.G.K. and A.A.P. contributed experimentally.
M.R.H. and D.E. synthesized the compounds. J.F.A., B.M.D.C.G., C.M.F., A.H.C., R.M.K., H.L.G.E., R.P.M., N.C.B., S.M.J., M.J.G. and M.S.E. carried out large-animal studies. G.G. and C.M.
provided naive nonhuman primate samples. J.F.A., B.M.D.C.G., M.R.H., C.M.F. and A.K. wrote the manuscript. CORRESPONDING AUTHOR Correspondence to Anastasia Khvorova. ETHICS DECLARATIONS
COMPETING INTERESTS A.K., J.F.A., M.R.H. and B.M.D.C.G. have filed a patent application for branched oligonucleotides. ADDITIONAL INFORMATION PUBLISHER’S NOTE: Springer Nature remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations. SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION Supplementary Figures 1–21, Supplementary
Tables 1 and 2 and Supplementary Notes 1 and 2 REPORTING SUMMARY RIGHTS AND PERMISSIONS Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Alterman, J.F., Godinho, B.M.D.C.,
Hassler, M.R. _et al._ A divalent siRNA chemical scaffold for potent and sustained modulation of gene expression throughout the central nervous system. _Nat Biotechnol_ 37, 884–894 (2019).
https://doi.org/10.1038/s41587-019-0205-0 Download citation * Received: 25 October 2018 * Accepted: 27 June 2019 * Published: 02 August 2019 * Issue Date: August 2019 * DOI:
https://doi.org/10.1038/s41587-019-0205-0 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not
currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative