Elsevier

Brain Research

Volume 1338, 18 June 2010, Pages 112-121
Brain Research

Review
RNAi therapeutics for CNS disorders

https://doi.org/10.1016/j.brainres.2010.03.038Get rights and content

Abstract

RNA interference (RNAi) is a process of sequence-specific gene silencing and serves as a powerful molecular tool to manipulate gene expression in vitro and in vivo. RNAi technologies have been applied to study gene function and validate drug targets. Researchers are investigating RNAi-based compounds as novel therapeutics to treat a variety of human diseases that are currently lacking sufficient treatment. To date, numerous studies support that RNAi therapeutics can improve disease phenotypes in various rodent models of human disease. Here, we focus on the development of RNAi-based therapies aimed at treating neurological disorders for which reduction of mutant or toxic gene expression may provide clinical benefit. We review RNAi-based gene-silencing strategies, proof-of-concept studies testing therapeutic RNAi for CNS disorders, and highlight the most recent research aimed at transitioning RNAi-based therapeutics toward clinical trials.

Introduction

Treatment of neurological diseases affecting the central nervous system (CNS) has proven to be a major challenge for clinicians. As average life span continues to increase among the human population, so does the societal burden caused by age-related neurodegenerative conditions; the two most prominent being Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases, among others, may have known genetic components or may appear sporadically with unknown etiology. By contrast, some neurological disorders [e.g., Huntington's disease (HD) and several spinocerebellar ataxias (SCAs)] are solely caused by the inheritance of genetic mutations. In recent years, there has been considerable progress made in elucidating the pathogenic mechanisms underlying these various neurological diseases; however, there are currently no cures, and therapies are largely symptomatic. Thus, researchers are investigating innovative therapeutic strategies to treat these diseases. One such approach is to silence (i.e., turn off or reduce) the expression of genes that cause or contribute to disease phenotypes. For neurological conditions with more complex origins, the candidate target genes are often less clear; however, gene-silencing strategies may be employed to inhibit cellular pathways that contribute to disease manifestation. For several autosomal dominant neurodegenerative diseases, gene mapping has identified the disease-causing mutations, facilitating candidate target gene selection for therapeutic silencing. Notably, in some cases, researchers have validated therapeutic target genes using tetracycline-regulated transgenic mouse models of dominant neurodegenerative diseases. These inducible models—in which the expression of mutant genes can be turned on or off—serve as powerful tools for assessing the reversibility of neurological conditions and evaluating disease-causing genes as therapeutic targets. For example, using these models, independent groups working on HD and SCA1 demonstrated that neuropathological and abnormal behavioral features of disease developed over time when the respective mutant proteins were expressed (Yamamoto et al., 1984, Zu et al., 2004). However, when transgene expression was turned off in affected mice, disease progression halted and pathological and behavioral features improved. Together, these experiments serve as proof-of-principle studies supporting the notion that inhibiting the expression of disease-causing genes may provide therapeutic benefit in patients already exhibiting disease phenotypes. In recent years, scientists have been rigorously investigating a variety of strategies to selectively inhibit gene expression with high specificity. To date, RNA interference (RNAi), which is capable of gene-specific targeting of messenger RNAs (mRNAs), has shown beneficial effects in cell and animal models.

Section snippets

RNAi overview

RNAi is a natural cellular process that serves to regulate gene expression and provide an innate defense mechanism against viral invasion and transposable elements (McManus and Sharp, 2002). The identification of the RNAi process has been recognized among the most significant contributions to cell biology. In 2006, the Nobel Prize in Physiology or Medicine was awarded to researchers Craig Mellow and Andrew Fire for their crucial role in the discovery of RNAi (Fire et al., 1998). Having been

Tools for RNAi

With a better understanding of endogenous miRNA biogenesis and gene silencing processes, scientists have devised strategies to co-opt the RNAi machinery to specifically silence various genes of interest. In this way, RNAi serves as a powerful molecular tool used to study gene function in biological processes and provides a novel strategy to treat a variety of diseases (e.g., dominant genetic disorders, cancer, and viral invasion, among others). The application of RNAi as a biological or

Proof-of-concept studies testing therapeutic RNAi in the CNS

During the past decade, scientists have made much progress in evaluating RNAi-based therapeutics for diseases affecting the CNS (Table 1). The potential of RNAi therapies has been tested in several cell and animal models of human neurodegenerative disease. The polyglutamine (polyQ)-repeat disease family has been a major focus for testing RNAi-based therapeutics in several laboratories (Caplen et al., 2002, Harper et al., 2005, Kubodera et al., 2005, Li et al., 2004, Xia et al., 2006). There are

Transitioning RNAi toward the clinic

The success of proof-of-concept studies highlights the potential utility of RNAi-based therapeutics for neurological disorders. As a step toward transitioning these strategies to the clinic, experiments to assess the safety, delivery, and dosing of RNAi reagents have been performed. One of the major considerations for gene-silencing applications is specificity. Though many reports endorse the potency and specificity of RNAi-based approaches, the issue remains largely unresolved across tissues

Summary

Since the discovery of RNAi 10 years ago, researchers have made substantial progress in elucidating the machinery and mechanisms involved in this gene silencing pathway. Scientists have used RNAi-based technologies to silence numerous genes in a variety of cell culture and animal models. Encouraging results from several proof-of-concept studies highlight the potential that RNAi therapeutics have for treating numerous disorders, including neurological conditions. Although there are significant

References (104)

  • Y.S. Lee et al.

    Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways

    Cell

    (2004)
  • B.P. Lewis et al.

    Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets

    Cell

    (2005)
  • M.S. Lombardi et al.

    A majority of Huntington's disease patients may be treatable by individualized allele-specific RNA interference

    Exp. Neurol.

    (2009)
  • L. Mangiarini 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

    (1996)
  • M.A. Minks et al.

    Structural requirements of double-stranded RNA for the activation of 2′, 5′-oligo(A) polymerase and protein kinase of interferon-treated HeLa cells

    J. Biol. Chem.

    (1979)
  • S. Pebernard et al.

    Determinants of interferon-stimulated gene induction by RNAi vectors

    Differentiation

    (2004)
  • E.L. Pfister et al.

    Five siRNAs targeting three SNPs may provide therapy for three-quarters of Huntington's disease patients

    Curr. Biol.

    (2009)
  • E. Rodriguez-Lebron et al.

    Intrastriatal rAAV-mediated delivery of anti-huntingtin shRNAs induces partial reversal of disease progression in R6/1 Huntington's disease transgenic mice

    Mol. Ther.

    (2005)
  • E. Rodriguez-Lebron et al.

    Allele-specific RNAi mitigates phenotypic progression in a transgenic model of Alzheimer's disease

    Mol. Ther.

    (2009)
  • M.K. Sapru et al.

    Silencing of human alpha-synuclein in vitro and in rat brain using lentiviral-mediated RNAi

    Exp. Neurol.

    (2006)
  • F. Saudou et al.

    Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions

    Cell

    (1998)
  • G. Schaffar et al.

    Cellular toxicity of polyglutamine expansion proteins: mechanism of transcription factor deactivation

    Mol. Cell.

    (2004)
  • D.S. Schwarz et al.

    Asymmetry in the assembly of the RNAi enzyme complex

    Cell

    (2003)
  • A. Ulusoy et al.

    Dose optimization for long-term rAAV-mediated RNA interference in the nigrostriatal projection neurons

    Mol. Ther.

    (2009)
  • Y.L. Wang et al.

    Clinico-pathological rescue of a model mouse of Huntington's disease by siRNA

    Neurosci. Res.

    (2005)
  • H. Wang et al.

    Therapeutic gene silencing delivered by a chemically modified small interfering RNA against mutant SOD1 slows amyotrophic lateral sclerosis progression

    J. Biol. Chem.

    (2008)
  • X. Xia et al.

    Allele-specific RNAi selectively silences mutant SOD1 and achieves significant therapeutic benefit in vivo

    Neurobiol. Dis.

    (2006)
  • T. Yamamoto et al.

    The human LDL receptor: a cysteine-rich protein with multiple Alu sequences in its mRNA

    Cell

    (1984)
  • A. Agrawal et al.

    Functional delivery of siRNA in mice using dendriworms

    ACS Nano.

    (2009)
  • V.A. Alvarez et al.

    Retraction of synapses and dendritic spines induced by off-target effects of RNA interference

    J. Neurosci.

    (2006)
  • S. Alves et al.

    Allele-specific RNA silencing of mutant ataxin-3 mediates neuroprotection in a rat model of Machado-Joseph disease

    PLoS ONE

    (2008)
  • A. Birmingham et al.

    3′ UTR seed matches, but not overall identity, are associated with RNAi off-targets

    Nat. Methods

    (2006)
  • D. Boden et al.

    Enhanced gene silencing of HIV-1 specific siRNA using microRNA designed hairpins

    Nucleic Acids Res.

    (2004)
  • A.J. Bridge et al.

    Induction of an interferon response by RNAi vectors in mammalian cells

    Nat. Genet.

    (2003)
  • X. Cai et al.

    Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs

    Rna

    (2004)
  • N.J. Caplen et al.

    Rescue of polyglutamine-mediated cytotoxicity by double-stranded RNA-mediated RNA interference

    Hum. Mol. Genet.

    (2002)
  • J.T. Chi et al.

    Genomewide view of gene silencing by small interfering RNAs

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • K.H. Chung et al.

    Polycistronic RNA polymerase II expression vectors for RNA interference based on BIC/miR-155

    Nucleic Acids Res.

    (2006)
  • B.L. Davidson et al.

    Viral vectors for gene delivery to the nervous system

    Nat. Rev. Neurosci.

    (2003)
  • M. DiFiglia 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)
  • V. Drouet et al.

    Sustained effects of nonallele-specific Huntingtin silencing

    Ann. Neurol.

    (2009)
  • S.M. Elbashir et al.

    RNA interference is mediated by 21- and 22-nucleotide RNAs

    Genes Dev.

    (2001)
  • J.L. Eriksen et al.

    Molecular pathogenesis of Parkinson disease

    Arch. Neurol.

    (2005)
  • W.P. Esler et al.

    A portrait of Alzheimer secretases—new features and familiar faces

    Science

    (2001)
  • Y. Fedorov et al.

    Off-target effects by siRNA can induce toxic phenotype

    RNA

    (2006)
  • A. Fire et al.

    Potent and specific genetic interference by double-stranded RNA in caenorhabditis elegans

    Nature

    (1998)
  • R.J. Fish et al.

    Short-term cytotoxic effects and long-term instability of RNAi delivered using lentiviral vectors

    BMC Mol. Biol.

    (2004)
  • K. Forstemann et al.

    Normal microRNA maturation and germ-line stem cell maintenance requires Loquacious, a double-stranded RNA-binding domain protein

    PLoS Biol.

    (2005)
  • K.D. Foust et al.

    Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes

    Nat. Biotechnol.

    (2009)
  • C.S. Gondi et al.

    RNAi-mediated inhibition of cathepsin B and uPAR leads to decreased cell invasion, angiogenesis and tumor growth in gliomas

    Oncogene

    (2004)
  • Cited by (39)

    • Stimuli-responsive In situ gelling system for nose-to-brain drug delivery

      2020, Journal of Controlled Release
      Citation Excerpt :

      The potency of siRNA49 in the treatment of different brain disorders like AD, PD, glioma etc. is well studied in the past few years. Though, the delimited BBB permeation of such negatively charged, high molecular weight compound reduces its therapeutic efficacy [163]. Hence, Perez et al. 2012 worked on direct nose-to-brain delivery of siRNA, which bypasses the BBB and improves the concentration of bioactive in the brain.

    • Broad therapeutic benefit after RNAi expression vector delivery to deep cerebellar nuclei: Implications for spinocerebellar ataxia type 1 therapy

      2014, Molecular Therapy
      Citation Excerpt :

      Methods to accomplish gene silencing include RNA interference (RNAi),8,9,10,11 antisense oligonucleotides,12,13,14 and inhibitory antibodies.15,16 RNAi is an evolutionarily conserved process that induces posttranscriptional gene silencing17 and has been co-opted for therapeutic development to silence pathogenic gene targets, including gain-of-function central nervous system diseases.18,19 We showed earlier that RNAi triggers released from first-generation short hairpin RNAs10 or artificial microRNA platforms20 were therapeutic in SCA1 transgenic mice.

    • Non-Viral nanosystems for gene and small interfering RNA delivery to the central nervous system: Formulating the solution

      2013, Journal of Pharmaceutical Sciences
      Citation Excerpt :

      Gene therapy can be broadly defined as the application of nucleic acids to express or silence genes in the treatment of disease.1–3 Gene therapies offer great potential as therapeutic approaches for neurological and neurodegenerative diseases, many of which lack satisfactory treatments.4,5 This includes the use of plasmid DNA (pDNA) encoding a functional therapeutic gene, or the use of either pDNA expressing short hairpin RNA (shRNA) or synthetic siRNAs (small interfering RNAs) to inhibit target gene expression.1,2,6

    • An in silico approach to design potential siRNA molecules for ICP22 (US1) gene silencing of different strains of human herpes simplex 1

      2013, Journal of Young Pharmacists
      Citation Excerpt :

      This finding is helpful to meet the demand of same treatment against different strains of same virus or against different viruses. The advance approach, RNAi technology for post-transcriptional silencing of specific gene is successfully adopted in several cases such as colorectal cancer therapeutics, therapeutics of hepatitis C virus,16 for CNS disorder,17 siRNA therapeutics can now possible in metabolic diseases of liver and are being evaluated in clinical trials for hypercholesterolemia.18 Synthetic siRNAs targeted against the viral structural Env proteins produced by HIV-1 can specifically suppress the expression of HIV-1 genes.

    • The role of IκB kinase complex in the neurobiology of Huntington's disease

      2011, Neurobiology of Disease
      Citation Excerpt :

      Thus, strategies should be developed to selectively inhibit IKKβ in the brain. An interesting therapeutic strategy is to knockdown the expression of IKKβ in the CNS by delivery of shRNAs, synthetic miRNAs, or anti-sense oligonucleotides (Boudreau and Davidson, 2010). It is notable that deleting IKKβ in the CNS of mice has no visible effect on neurodevelopment, growth, and indeed ameliorates neurodegeneration in ischemia and MS models (Herrmann et al., 2005; Van Loo et al., 2006).

    View all citing articles on Scopus
    View full text