Interfering with the brain: Use of RNA interference for understanding the pathophysiology of psychiatric and neurological disorders

https://doi.org/10.1016/j.pharmthera.2005.08.006Get rights and content

Abstract

Psychiatric and neurological disorders are among the most complex, poorly understood, and debilitating diseases in medicine. The burgeoning advances in functional genomic technologies have led to the identification of a vast number of novel genes that are potentially implicated in the pathophysiology of such disorders. However, many of these candidate genes have not yet been functionalized and require validation in vivo. Traditionally, abrogating gene function is one of the primary means of examining the physiological significance of a given gene product. Several methods have been developed for gene ablation or knockdown, however, with limited levels of success. The recent discovery of RNA interference (RNAi), as a highly efficient method for gene knockdown, has been one of the major breakthroughs in molecular medicine. In vivo application of RNAi is further demonstrating the promise of this technology. Recent efforts have focused on applying RNAi-based knockdown to understand the genes implicated in neuropsychiatric disorders. However, the greatest challenge with this approach is translating the success of RNAi from mammalian cell cultures to the brain in animal models of disease and, subsequently, in patients. In this review, we describe the various methods that are being developed to deliver RNAi into the brain for down-regulating gene expression and subsequent phenotyping of genes in vivo. We illustrate the utility of various approaches with a few successful examples and also discuss the potential benefits and pitfalls associated with the use of each delivery approach. Appropriate tailoring of tools that deliver RNAi in the brain may not only aid our understanding of the complex pathophysiology of neuropsychiatric disorders, but may also serve as a valuable therapy for disorders, where there is an immense unmet medical need.

Introduction

Perowne: “Best hope now, apparently, is RNA interference.” Baxter: “Yeah. Gene silencing. One day, perhaps…” From “Saturday” by Ian McEwan, Jonathan Cape Press, 2005.

Since its recent discovery, first in the worm, Caenorhabditis elegans (Fire et al., 1998), and subsequently in mammalian cells (Bitko & Barik, 2001, Caplen et al., 2001, Elbashir et al., 2001a), RNA interference (RNAi) has emerged as one of the most important innovations in modern molecular medicine. This is reflected by the fact that the discovery of small RNAs was Science magazine's “Scientific Breakthrough of the Year” in 2002 (Couzin, 2002). A number of biotechnology companies have already been established, with strategies specifically dedicated to develop RNAi-based medicines in order to cure diseases that have been traditionally difficult to approach (Pollack, 2004). This also led the Fortune magazine to label RNAi as “Biotech's Billion Dollar Breakthrough” in 2003 (Stipp, 2003). The ascent of RNAi is further revealed in that it has already permeated the realms of modern literary fiction, as in Ian McEwan's latest novel. In this review, we focus on the potential utility of RNAi to impact our understanding of the brain function. It is hoped that RNAi-based in vivo methods will improve the target validation process for neuropsychiatric disorders. Further, we describe how such efforts may lead to the development of better therapeutic strategies for psychiatric and neurodegenerative diseases where there is an immense unmet medical need.

Post-genomic biomedical research has paved our way from mere clinical observations of neuropsychiatric disorders to modern microarray strategies that help uncover disease mechanisms and detect potentially target genes relevant to the disease pathology (Mirnics et al., 2000, Dickey et al., 2003, McClung & Nestler, 2003, Newton et al., 2003, Wong & Licinio, 2004, Yao et al., 2004). Microarray-based genomic analyses are further complimented by proteomic platforms that enable the identification of proteins with altered expression in a disease state (Denslow et al., 2003, Rohlff & Hollis, 2003, Freeman & Hemby, 2004, Kim et al., 2004b, Vercauteren et al., 2004, Williams et al., 2004, Zhang & Goodlett, 2004). Consequently, the high-throughput gene and protein expression profiling permit the systematic identification of a large number of targets, or so-called “hits”, with pathophysiological implications or relevance. Targets identified from the use of such high-throughput technologies warrant an in vivo validation of their etiological role in the disease.

In vivo validation of targets is currently accomplished using tools that employ either pharmacological, immunological, or genetic means of manipulating the target function (Fig. 1). Pharmacological tools directly activate (e.g., agonists, positive allosteric modulators) or inhibit (e.g., antagonists, negative allosteric modulators) the protein product of the gene and provide a rapid means of in vivo target validation. Similarly, antibodies serve as immunological tools that readily inhibit the target protein to validate its function in vivo (Salfeld, 2004). However, the rapidity of these tools in revealing a target-based phenotype is largely superseded by both the difficulty and time-consuming process of finding small molecule compounds or generating antibodies that are selective for the target protein. Too often, we lack highly selective and potent ligands for even well-known targets. Another disadvantage of the pharmacological approach for initial target validation, following gene microarray studies, is that the gene product must be identified and reasonably well characterized. This is not always the case with genetic studies, where many of the hits are genes, which have not been functionalized or are not full-length sequences but just expressed sequence tags (ESTs).

The genetic approach of producing target mutants offers an alternative far selective to the pharmacological or immunological means of target validation in vivo. It includes genetic manipulation by homologous recombination in the embryonic stem (ES) cells, such that the gene of interest is either overexpressed or knocked out to produce stable transgenic animals (Capecchi, 1989, Koller & Smithies, 1992, Shastry, 1998). However, a major disadvantage to the use of this methodology is the restricted feasibility of homologous recombination to rodent ES cells, which is further compounded by the phenotypic differences observed among different strains following mutation of the same gene (Crawley, 1996, Gerlai, 1996, Lathe, 1996, Crabbe et al., 1999, Nadeau, 2001). In addition to the enormous amount of time involved in generating stably expressing mutant offsprings, this approach is not applicable to the phenotyping of genes that are critically involved in the early developmental stages of life. Furthermore, the most significant and commonly encountered drawback of producing mutant animals is the genetic compensation as well as developmental adaptations that may mask the establishment of a clear phenotype. Generation of inducible transgenics evades several limitations associated with the use of traditional mutant animals (van der Neut, 1997, Lewandoski, 2001, Misra & Duncan, 2002, Williams et al., 2003, Beglopoulos & Shen, 2004). Nonetheless, this process still remains time and labor intensive, which provides further impetus for research efforts into the development of other more efficient ways of manipulating gene expression during adulthood.

Manipulating the gene expression in somatic systems circumvents some of the problems encountered with transgenic mutants. In this case, gene knockdown is achieved using antisense oligodeoxynucleotides (Robinson et al., 1997, Weiss et al., 1997, Szklarczyk & Kaczmarek, 1999, Godfray & Estibeiro, 2003) or ribozymes (Welch et al., 1998, Cairns et al., 2002); however, at unpredictable frequencies and occasional acquaintance of non-specific events. Clearly, the shortcomings of existing methodologies demand an effective alternative that enables a selective and rapid validation of novel targets in vivo. More recently, the endogenous regulatory mechanism of RNAi has emerged as a potentially superior alternative to the traditional approaches for target validation in adult animals.

Section snippets

RNA interference

RNAi is a cellular surveillance phenomenon that serves to not only repress viral infections, transposable elements, and repetitive genes, but also to regulate gene expression as well as normal cell development (Ambros, 2004, Bender, 2004, Ding et al., 2004, Lippman & Martienssen, 2004, Schramke & Allshire, 2004). Although originally described in the flowering plant petunia (Napoli et al., 1990, van der Krol et al., 1990), it was the discovery of RNAi mechanisms in the worm, C. elegans, by Fire

RNAi in the brain

An increasing number of studies in model organisms, such as C. elegans (Poinat et al., 2002, Bianchi et al., 2003, Schulze et al., 2003, Henricson et al., 2004, Nollen et al., 2004, Withee et al., 2004, Jiang et al., 2005) and Drosophila melanogaster (Mihaly et al., 2001, Ishizuka et al., 2002, Ozon et al., 2002, Chan et al., 2003, Dzitoyeva et al., 2003, Yang et al., 2003, Dunlop et al., 2004, Geng et al., 2004, Rival et al., 2004, Xu et al., 2004), demonstrate the effective use of RNAi in

Conclusions

Since its revelation in the mammalian systems in 2001, RNAi has rapidly emerged as a tool far more selective and robust than the currently available approaches for analyses of gene function. RNAi has, by now, revolutionized the world of functional genomics (Dorsett & Tuschl, 2004, Silva et al., 2004), and its translation from in vitro to in vivo holds the key to its successful application in the target validation process. Delivery of RNAi to the brain stages a crucial factor in its in vivo use

Acknowledgments

We gratefully acknowledge Drs. Kumlesh K. Dev and Klemens Kaupmann for their helpful discussions and critical review of the manuscript.

References (346)

  • E.N. Burright et al.

    SCA1 transgenic mice: a model for neurodegeneration caused by an expanded CAG trinucleotide repeat

    Cell

    (1995)
  • E. Butkevich et al.

    Drebrin is a novel connexin-43 binding partner that links gap junctions to the submembrane cytoskeleton

    Curr Biol

    (2004)
  • M.M. Cardenas et al.

    STK, the src homologue, is responsible for the initial commitment to develop head structures in Hydra

    Dev Biol

    (2003)
  • V.A. Colotla et al.

    Effects of MPTP on locomotor activity in mice

    Neurotoxicol Teratol

    (1990)
  • J.N. Crawley

    Unusual behavioral phenotypes of inbred mouse strains

    Trends Neurosci

    (1996)
  • J.F. Cryan et al.

    Non-nicotinic neuropharmacological strategies for nicotine dependence: beyond bupropion

    Drug Discov Today

    (2003)
  • T. Dalmay et al.

    An RNA-dependent RNA polymerase gene in Arabidopsis is required for posttranscriptional gene silencing mediated by a transgene but not by a virus

    Cell

    (2000)
  • B.L. Davidson et al.

    Viral delivery of recombinant short hairpin RNAs

    Methods Enzymol

    (2005)
  • W. Davies

    Knockdown not knockout

    Drug Discov Today

    (2005)
  • S.W. Ding et al.

    RNA silencing: a conserved antiviral immunity of plants and animals

    Virus Res

    (2004)
  • G. Dityateva et al.

    Rapid and efficient electroporation-based gene transfer into primary dissociated neurons

    J Neurosci Methods

    (2003)
  • J. Dunlop et al.

    Glaikit is essential for the formation of epithelial polarity and neuronal development

    Curr Biol

    (2004)
  • F. Francis et al.

    Doublecortin is a developmentally regulated, microtubule-associated protein expressed in migrating and differentiating neurons

    Neuron

    (1999)
  • L. Gan et al.

    Specific interference with gene expression and gene function mediated by long dsRNA in neural cells

    J Neurosci Methods

    (2002)
  • L. Gan et al.

    Identification of cathepsin B as a mediator of neuronal death induced by Abeta-activated microglial cells using a functional genomics approach

    J Biol Chem

    (2004)
  • S. Ge et al.

    Caveolin-1 knockdown by small interfering RNA suppresses responses to the chemokine monocyte chemoattractant protein-1 by human astrocytes

    J Biol Chem

    (2004)
  • J. Gehl et al.

    In vivo electroporation of skeletal muscle: threshold, efficacy and relation to electric field distribution

    Biochim Biophys Acta

    (1999)
  • J. Gehl et al.

    Vascular reactions to in vivo electroporation: characterization and consequences for drug and gene delivery

    Biochim Biophys Acta

    (2002)
  • C. Geng et al.

    Complete RNAi rescue of neuronal degeneration in a constitutively active Drosophila TRP channel mutant

    Biochim Biophys Acta

    (2004)
  • R. Gerlai

    Gene-targeting studies of mammalian behavior: is it the mutation or the background genotype?

    Trends Neurosci

    (1996)
  • H. Giladi et al.

    Small interfering RNA inhibits hepatitis B virus replication in mice

    Mol Ther

    (2003)
  • A. Abdelgany et al.

    Allele-specific silencing of a pathogenic mutant acetylcholine receptor subunit by RNA interference

    Hum Mol Genet

    (2003)
  • L. Aigner et al.

    Isolated lissencephaly sequence and double-cortex syndrome in a German family with a novel doublecortin mutation

    Neuropediatrics

    (2000)
  • Y. Akaneya et al.

    RNAi-induced gene silencing by local electroporation in targeting brain region

    J Neurophysiol

    (2005)
  • L. Alberi et al.

    Engrailed genes are cell-autonomously required to prevent apoptosis in mesencephalic dopaminergic neurons

    Development

    (2004)
  • C.R. Allerson et al.

    Fully 2′-modified oligonucleotide duplexes with improved in vitro potency and stability compared to unmodified small interfering RNA

    J Med Chem

    (2005)
  • S.G. Amara et al.

    Neurotransmitter transporters: recent progress

    Annu Rev Neurosci

    (1993)
  • M. Amarzguioui et al.

    Tolerance for mutations and chemical modifications in a siRNA

    Nucleic Acids Res

    (2003)
  • V. Ambros

    The functions of animal microRNAs

    Nature

    (2004)
  • M.S. Ansorge et al.

    Early-life blockade of the 5-HT transporter alters emotional behavior in adult mice

    Science

    (2004)
  • A. Asakawa et al.

    Effects of agouti-related protein, orexin and melanin-concentrating hormone on oxygen consumption in mice

    Int J Mol Med

    (2002)
  • J. Bai et al.

    RNAi reveals doublecortin is required for radial migration in rat neocortex

    Nat Neurosci

    (2003)
  • I. Bantounas et al.

    Assessing adenoviral hammerhead ribozyme and small hairpin RNA cassettes in neurons: inhibition of endogenous caspase-3 activity and protection from apoptotic cell death

    J Neurosci Res

    (2005)
  • S. Battersby et al.

    Structure of a variable number tandem repeat of the serotonin transporter gene and association with affective disorder

    Psychiatr Genet

    (1996)
  • D. Baulcombe

    RNA silencing in plants

    Nature

    (2004)
  • G. Beale et al.

    Gene silencing nucleic acids designed by scanning arrays: anti-EGFR activity of siRNA, ribozyme and DNA enzymes targeting a single hybridization-accessible region using the same delivery system

    J Drug Target

    (2003)
  • V. Beglopoulos et al.

    Gene-targeting technologies for the study of neurological disorders

    Neuromol Med

    (2004)
  • E. Bernstein et al.

    Role for a bidentate ribonuclease in the initiation step of RNA interference

    Nature

    (2001)
  • A. Bhargava et al.

    Glucocorticoids prolong Ca(2+) transients in hippocampal-derived H19-7 neurons by repressing the plasma membrane Ca(2+)-ATPase-1

    Mol Endocrinol

    (2002)
  • D.H. Bhatt et al.

    Cyclic AMP-induced repair of zebrafish spinal circuits

    Science

    (2004)
  • Cited by (65)

    • Oligonucleotides as therapeutic tools for brain disorders: Focus on major depressive disorder and Parkinson's disease

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      However, burgeoning advances in functional genomic technologies have led to the identification of a vast number of novel genes that are potentially implicated in the MDD pathophysiology. In this way, several key genes linked to serotonergic neurons, neurogenesis, neuronal plasticity, synaptogenesis, and transcription factors, as well as cytokine signaling and glucocorticoid receptor signaling, among others, have been involved in MDD (Lin & Tsai, 2019; Thakker, Hoyer, & Cryan, 2006). Many of these candidate genes have not yet been functionalized and require validation in vivo.

    • Cyclodextrins based delivery systems for macro biomolecules

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      The cell viability assay and cellular uptake studies of PR+ nanoparticles compared to bPEI displayed 100 to 200 times less toxic and even better cellular uptake percentage during the N/P ratio was 20 [144]. In order to overcome the issues in delivering siRNA into neurons and the central nervous system, like neuronal uptake, vesicular escape, and blood-brain barrier et al. [145,146] to treat some nervous system diseases, O’Driscoll and Cryan teams collaborated to devise a neuronal siRNA delivery carrier based on modified β-CD [139]. They introduced lipophilic groups on the primary face and the cationic groups via “click chemistry” on the secondary face of β-CD selectively to form a functionalized CD monomer [111] (See Fig. 22).

    • Delivering a disease-modifying treatment for Huntington's disease

      2015, Drug Discovery Today
      Citation Excerpt :

      Additionally, not only because of the sequence mismatch and partial complementarity, but also through the ability of binding to several UTR sequences, miRNAs have been shown to silence several mRNA targets [38]. This RNAi machinery can be artificially hijacked to induce specific gene expression knockdown, which is commonly performed using synthetic siRNAs, not only in combination with nonviral delivery systems, but also through shRNA, usually utilising viral delivery systems [42–44]. After delivery, shRNAs are expressed within the nucleus and need to be transported by exportin-5 to the cytoplasm, whereas synthetic siRNAs bypass this nuclear step.

    • 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 :

      Such typical non-viral vectors are described in Table 1, along with some of their advantages and disadvantages. Delivery to the CNS is particularly challenging because of its anatomical and physiological complexities,28,29 as illustrated in Figure 1. The CNS is protected by the blood–brain barrier (BBB), which consists of tightly joined capillary endothelial cells, restricting access of large molecules into the brain30 (Fig. 1a).

    View all citing articles on Scopus
    1

    Present address: Biosciences R&D, Medtronic, Inc., Minneapolis, MN 55432, USA.

    View full text