ArticlePDF Available
RNA interference: research tool
or therapeutic technique?
Editorial
Correspondence:
Eleni Tzortzaki MD, PhD
Lecturer in Thoracic Medicine
Department of Thoracic Medicine,
University Hospital of Heraklion
Medical School, University of Crete
71110 Heraklion, Crete, Greece
Tel: (30) 2810 392433, Fax: (30) 2810542650
E-mail: tzortzaki@med.uoc.gr
RNA interference (RNAi) represents a fundamental biological process by
which cells regulate gene silencing post-transcriptionally. It is triggered by
double-stranded RNA (dsRNAs) precursors that vary in length and origin.
These dsRNAs are rapidly processed into short RNA duplexes of about 21 to
28 nucleotides in length, which then guide the recognition and ultimately
the cleavage or translational repression of complementary single-stranded
RNAs. RNAi targets include RNA from viruses and transposons (significant
for some forms of innate immune response), and they also play a role in
regulating development and genome maintenance (since short RNAs have
been implicated in guiding chromatin modification)1.
SMALL RNAs
Small interfering RNA strands (siRNA) are RNA sequences, roughly 23
nucleotides in length, which are crucial regulators of gene expression. They
have nucleotide sequences complementary to the targeted messenger RNA
strand, mediating mRNA degradation and or/ repressing the translation
of the mRNA into protein2. Specific RNAi pathway proteins are guided by
the siRNA to the targeted messenger RNA (mRNA), where they “cleave”
the target, breaking it down into smaller portions that can no longer be
translated into protein. A type of RNA transcribed from the genome itself,
microRNA (miRNA), works in the same way. In mammals, three major classes
of small regulatory RNAs have been identified so far: microRNAs (miRNAs),
short-interfering RNAs (shRNAs), and Piwi-assosiated RNAs (piRNAs). The
precursors of the miRNAs and piRNAs are endogenously encoded primary
RNA transcripts, while shRNAs are produced by the cleavage of exogenous
dsRNAs2.
RNAi PATHwAY
RNAi is an RNA-dependent gene silencing process that is controlled by
the RNA-induced silencing complex (RISC) and is initiated by short double-
stranded RNA molecules in the cytoplasm of a cell, where they interact with
the catalytic RISC component Αrgonaute1. Despite the differences in the
size and biogenesis of these small regulatory RNAs, their RNAi interference
Eleni G. Tzortzaki1,2,
Maria Psarrou2,
Nikolaos M. Siafakas1,2
1Department of Thoracic Medicine, Medical School,
University of Crete, Greece
2Laboratory of Molecular and Cellular Pneumonol-
ogy, Medical School, University of Crete, Greece
Key words:
siRNA,
miRNA,
RNAi,
Lung,
Gene-silencing
-
-
-
-
-
236 ΠΝΕΥΜΩΝ Τεύχος 3ο, Τόμος 21ος, Ιούλιος - Σεπτέμβριος 2008
pathways involve proteins that belong to the Argonaute
family
3
. These proteins make up a highly conserved
family whose members have been implicated in RNAi
and related phenomena in several organisms. In addition
to roles in RNAi-like mechanisms, Argonaute proteins
(AGO) influence development, and at least one subset
is involved in stem cell fate determination3.
The RNAi pathway is initiated by the enzyme Dicer,
which cleaves long, double-stranded dsRNA molecules
into short fragments of 20–25 base pairs. One of the
two strands of each fragment, known as the guide
strand, is then incorporated into the RISC and pairs with
complementary sequences
1,4
. More specifically, in an
initiation step, exogenous (e.g., viral) ds-RNA is digested
by the enzyme Dicer, a member of the RNase III family of
ds-RNA-specific ribonucleases, into siRNAs. In the effector
step, the antisense strand of siRNAs is incorporated into
the RISC, which then targets the mRNA complementary
to the siRNA within the complex, cleaves the target
mRNA, thus leading to inhibition of mRNA translation
(Figure 1A). In mammalian cells, RNAi can begin with
endogenously encoded primary micro-RNA transcripts
(pri-miRNA) that are transcribed by polymerase II (Pol II).
The precursor miRNA is bound in the cytoplasm by the
Dicer complex which processes it further for loading into
the AGO2-RISC complex. The AGO2-RISC-bound miRNA
then recognizes target sites in the mRNA, leading to
translation repression, hence halting production of the
corresponding protein (Figure 1B)4.
siRNA may be obtained by chemical synthesis or
expressed from a DNA-vector. In the latter process, a
DNA insert of about 70 bp encoding a short hairpin RNA
Figure 1. RNA interference (RNAi) pathways involve either small interfering RNA (siRNA) or micro-RNA (miRNA). A. The siRNA
pathway involves cleavage of long double-stranded RNA (dsRNA) by the Dicer enzyme complex to yield siRNA. These siRNA are
then loaded into Argonaute 2 (AGO2) and the RNAi-induced silencing complex (RISC). The active RISC with guide strand recog-
nizes target sites on mRNA and performs site-specific cleavage of the mRNA. B. The miRNA pathway begins with endogenously
encoded primary micro-RNA transcripts (pri-miRNA) that are transcribed by polymerase II (Pol II). The precursor miRNA is bound
in the cytoplasm by the Dicer complex which processes it further for loading into the AGO2-RISC complex. The AGO2-RISC-bound
miRNA then recognizes target sites in the mRNA, leading to translation repression. Modified from de Fougerolles A & Novobrantseva
T, Curr Opin Pharm 2008 (16).
237PNEUMON Number 3, Vol. 21, July - September 2008
(shRNA) targeting the gene of interest is cloned into an
expression vector. When the insert-containing vector is
transfected into the cell, it expresses the shRNA which is
rapidly processed by the cellular machinery into a 19–22
nucleotide ds-RNA5.
BIOLOGICAL APPLICATIONS
RNAi is viewed as one of the most important recent dis-
coveries in biology
6
. Because RNAi confers transient inter-
ference of gene expression in a sequence-specific manner,
it represents a new class of nucleic acid-based molecules
likely to have significant medical utility. Identification of
potentially potent lead siRNA candidates usually begins
with bioinformatic design and siRNA synthesis, followed
by in vitro characterization of siRNA for potency and
specificity, which ultimately results in the selection of
siRNA drug candidates There are two major considera-
tions with regard to siRNA specificity: off-targeting’ due
to silencing of genes sharing partial homology with the
siRNA, and ‘immune stimulation’ due to the engage-
ment of components of the innate immune system by
the siRNA duplex7.
Having identified potentially optimized siRNA with
drug-like properties, the major hurdle remains of ef-
fective delivery to the target cell. The RNAi mechanism
has two important implications for drug discovery. First,
since RNAi-based gene silencing occurs in the cytoplasm,
where the RISC brings together the antisense strand of a
siRNA duplex and the corresponding mRNA molecule, any
method for delivery of siRNA-based therapeutics must be
capable of releasing the siRNA into the cytoplasm of the
relevant cells. This is the key challenge of siRNA delivery.
All other aspects of delivery, such as the targeting of
particular cell types, must be compatible with the basic
requirement that the cells must be able to take up the
siRNA and release it into the cytoplasm in an active form.
Second, since the process that leads to cleavage of the
mRNA strand does not consume the antisense strand of
the siRNA, the siRNA is able to act catalytically, and thus
can persist in silencing its gene for a significant period of
time without interfering with the genetic information8.
Several types of siRNA delivery are under active inves-
tigation, including: direct delivery of saline-formulated
siRNA; encapsulation into liposomes and lipoplexes;
conjugation to antibodies, peptides, aptamers, and other
molecules; and formation of complexes with chemical
and biological polymers. Each of these approaches has
potential advantages, but poses particular challenges8.
siRNA AND THE LUNG
As with any other nucleic acid, in the specific case of
delivery of siRNAs to the lung, the specific extracellular
barriers depend on the route of administration. Introduc-
tion of the complexes into the lung could be achieved,
in principle, by the intranasal, the intratracheal or the
systemic route8. The first two are particularly appealing
since they offer a unique opportunity for the delivery of
siRNAs to the airway and alveolar epithelium. However,
airway-directed gene delivery is not simple because the
lung has evolved both physical and immune barriers
that can hinder effective transduction of epithelial cells.
In addition to such physical phenomena as cilia beating
and mucociliary clearance, the possible interaction of the
complexes with the airway surface liquid (ASL) covering
the airway epithelial cells poses a further major barrier9.
First, negatively charged ASL constituents could bind
directly to positively charged complexes, altering their
size and switching their overall charge to negative, and
hence affecting their diffusion to the target cells or cel-
lular uptake. Additionally, binding of negatively charged
ASL components might displace the nucleic acid from
the complexes and consequently lower their delivery ef-
ficiency. Considering the foregoing difficulties, systemic
administration may be perceived as an alternative to
intranasal and intratracheal administration of the com-
plexes
8,9
. Owing to the similarities between siRNA and
DNA, considering the case of the complexes of DNA would
be instructive, the biodistribution of which is a complex
process dependent on their colloidal properties, as well
as their interaction with blood components. Furthermore,
scavenger cells usually absorb complexes bearing strong
anionic charge, which would result in elimination of the
genetic material from the body. A strong positive charge
on the complexes can also be deleterious. The distribu-
tion of transgene expression following administration of
DNA–polycation complexes is primarily observed in the
lungs and to a much lesser extent in other major organs,
such as the spleen, liver and heart. Transgene expression
is significantly altered when plasmid DNA is formulated
with polycations grafted with hydrophilic polymers, such
as pluronic. In this case, the expression of the reporter
gene is evenly distributed among the aforementioned
major organs. Similarly to the case of DNA, intravenous
injection of siRNA with PEG-containing liposomes has
also been reported to result in preferential delivery of
siRNA to the liver, implying that the surface properties
of the polyplexes is an important factor in determining
238 ΠΝΕΥΜΩΝ Τεύχος 3ο, Τόμος 21ος, Ιούλιος - Σεπτέμβριος 2008
their in vivo distribution9.
SiRNA could be rapidly designed to combat viral
targets. Local RNAi can protect against respiratory viral
infections including RSV
10
, PIV
10
, SARS-SCV
11
and influ-
enza
12,13
. Moreover, many endogenous lung targets have
been silenced in the context of acute lung injury (ALI)
models14,15. In summary, these reports suggest that, un-
like for systemic delivery, the direct administration of
saline-formulated siRNA under certain conditions is able
to silence specifically both viral and endogenous gene
targets expressed in lung epithelial cells10-15.
Delivery of siRNA directly to the lung has the advantage
that, as for any drug, the dose of siRNA required for efficacy
is substantially lower when administered near the target
cell type. In addition, direct delivery might also reduce
any theoretical undesired systemic side effects. Lastly,
and most important in the context of treating respiratory
disease, instillation of siRNA (for instance by intranasal or
intratracheal administration) could allow direct access to
lung epithelial cells in a variety of pulmonary disorders,
apart from viral infections, such as cystic fibrosis, chronic
obstructive pulmonary disease, asthma, and pulmonary
fibrosis16.
However, research is needed in order to formulate ways
of improving cellular uptake and cytoplasmic localization
of siRNA, to broaden the lung cell types to which siRNA
can be delivered, and to improve the pharmacokinetic
and lung distribution profile of siRNA. The rapid preclinical
progress and encouraging initial clinical results, suggest
that siRNA therapeutics may have a useful role in the
treatment of respiratory disease.
REFERENCES
1. Meister G, Tuschl T. Mechanisms of gene silencing by double-
stranded RNA. Nature 2004;431:343-9.
2. Mourelatos Z. Small RNAs: The seeds of silence. Nature
2008;455:44-5.
3. Carmell M, Xuan Z, Zhang MQ, Hannon GJ. The Argonaute
family: tentacles that reach into RNAi, developmental con-
trol, stem cell maintenance, and tumorigenesis. Genes Dev
2002;16:2733-42.
4. McManus MT, Sharp PA. Gene silencing in mammals by small
interfering RNAs, Nat Rev Genet 2002;3:737–47.
5. Wu MT, Wu RH, Hung CF, Cheng TL, Tsai WH, Chang WT. Simple
and efficient DNAvector based RNAi systems in mammalian
cells. Biochem Biophys Res Commun 2005;330:53–9.
6. Novina CD, Sharp PA. The RNAi revolution. Nature 2004;430:161–
4.
7. Bumcrot D, Manoharan M, Koteliansky V, Sah DW. RNAi thera-
peutics: a potential new class of pharmaceutical drugs. Nat
Chem Biol 2006;2:711-9.
8. Reidhaar-Olson JF. Delivering RNAi Therapeutics. Drug Discov
Dev 2008;11:22-5.
9. Thomas M, Lu JJ, Chen J, Klibanov AM. Non-viral siRNA delivery
to the lung. Adv Drug Deliver Rev 2007;59:124–33.
10. Bitko V, Musiyenko A, Shulyayeva O, Barik S. Inhibition of
respiratory viruses by nasally administered siRNA. Nat Med
2005;11:50-5.
11. Li BJ, Tang Q, Cheng D, et al. Using siRNA in prophylactic and
therapeutic regimens against SARS coronavirus in Rhesus
macaque. Nat Med 2005;11:944-51.
12. Ge Q, Filip L, Bai A, Nguyen T, Eisen HN, Chen J. Inhibition
of influenza virus production in virus-infected mice by RNA
interference. Proc Natl Acad Sci USA 2004;101:8676-81.
13. Tompkins SM, Lo CY, Tumpey TM, Epstein SL. Protection against
lethal influenza virus challenge by RNA interference in vivo.
Proc Natl Acad Sci USA 2004;101:8682-6.
14. Zhang X, Shan P, Jiang D, et al. Small interfering RNA targeting
heme oxygenase-1 enhances ischemia-reperfusion-induced
lung apoptosis. J Biol Chem 2004;279:10677-84.
15. Lomas-Neira JL, Chung CS, Wesche DE, Perl M, Ayala A. In vivo
gene silencing (with siRNA) of pulmonary expression of MIP-2
versus KC results in divergent effects on hemorrhage-induced,
neutrophil-mediated septic acute lung injury. J Leukoc Biol
2005;77:846-53.
16. de Fougerolles A, Novobrantseva T. siRNA and the lung: research
tool or therapeutic drug? Curr Opin Pharm 2008;8:280–285.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Among the 3 billion base pairs of the human genome, there are approximately 30,000-40,000 protein-coding genes, but the function of at least half of them remains unknown. A new tool - short interfering RNAs (siRNAs) - has now been developed for systematically deciphering the functions and interactions of these thousands of genes. siRNAs are an intermediate of RNA interference, the process by which double-stranded RNA silences homologous genes. Although the use of siRNAs to silence genes in vertebrate cells was only reported a year ago, the emerging literature indicates that most vertebrate genes can be studied with this technology.
Article
Full-text available
The term RNAi - short for RNA interference - crops up again and again in biology research these days. This is in part because of its power as a laboratory tool, and in part because it is a widespread natural phenomenon.
Article
Full-text available
Respiratory syncytial virus (RSV) and parainfluenza virus (PIV) are two respiratory pathogens of paramount medical significance that exert high mortality. At present, there is no reliable vaccine or antiviral drug against either virus. Using an RNA interference (RNAi) approach, we show that individual as well as joint infection by RSV and PIV can be specifically prevented and inhibited by short interfering RNAs (siRNAs), instilled intranasally in the mouse, with or without transfection reagents. The degree of protection matched the antiviral activity of the siRNA in cell culture, allowing an avenue for quick screening of an efficacious siRNA. When targeting both viruses in a joint infection, excess of one siRNA moderated the inhibitory effect of the other, suggesting competition for the RNAi machinery. Our results suggest that, if properly designed, low dosages of inhaled siRNA might offer a fast, potent and easily administrable antiviral regimen against respiratory viral diseases in humans.
Article
Full-text available
Lung injury in trauma patients exposed to a secondary infectious/septic challenge contributes to the high morbidity/mortality observed in this population. Associated pathology involves a dys-regulation of immune function, specifically, sequestration of activated polymorphonuclear neutrophils (PMN) in the lungs. The targeting of PMN is thought to involve the release of chemokines from cells within the local environment, creating a concentration gradient along which PMN migrate to the focus of inflammation. Keratinocyte-derived chemokine (KC) and macrophage-inflammatory protein-2 (MIP-2) are murine neutrophil chemokines identified as playing significant but potentially divergent roles in the pathogenesis of acute lung injury (ALI). In the current study, we examined the contribution of local pulmonary cells to the production of KC and MIP-2 and the pathogenesis of ALI. We hypothesized that local silencing of KC or MIP-2, via the local administration of small interference RNA (siRNA) against KC or MIP-2, following traumatic shock/hemorrhage (Hem), would suppress signaling for PMN influx to the lung, thereby reducing ALI associated with a secondary septic challenge (cecal ligation and puncture). Assessment of siRNA local gene silencing was done in green fluorescent protein (GFP)-transgenic, overexpressing mice. A marked suppression of GFP expression was observed in the lung 24 h following intratracheal (i.t.) instillation of GFP siRNA, which was not observed in the liver. To test our hypothesis, siRNA against KC or MIP-2 (75 ug/C3H/Hen mouse) was instilled (i.t.) 2 h post-Hem (35 mm Hg for 90 min, 4x LRS Rx.). Twenty-four hours after, mice were subjected to septic challenge and then killed 24 h later. i.t. MIP-2 siRNA significantly (P < 0.05, ANOVA-Tukey's test, n = 5-6/group) reduced tissue and plasma interleukin (IL)-6, tissue MIP-2 (enzyme-linked immunosorbent assay), as well as neutrophil influx [myeloperoxidase (MPO) activity]. In contrast, KC siRNA treatment reduced plasma KC, tissue KC, and IL-6 but produced no significant reduction in plasma IL-6 or MPO. Neither treatment reduced tissue or plasma levels of tumor necrosis factor alpha compared with vehicle. These data support not only our hypothesis that local pulmonary chemokine production of MIP-2, to a greater extent than KC, contributes to the pathogenesis of PMN-associated ALI following Hem but also the use of siRNA as a potential therapeutic.
Article
Full-text available
Development of therapeutic agents for severe acute respiratory syndrome (SARS) viral infection using short interfering RNA (siRNA) inhibitors exemplifies a powerful new means to combat emerging infectious diseases. Potent siRNA inhibitors of SARS coronavirus (SCV) in vitro were further evaluated for efficacy and safety in a rhesus macaque (Macaca mulatta) SARS model using clinically viable delivery while comparing three dosing regimens. Observations of SARS-like symptoms, measurements of SCV RNA presence and lung histopathology and immunohistochemistry consistently showed siRNA-mediated anti-SARS efficacy by either prophylactic or therapeutic regimens. The siRNAs used provided relief from SCV infection-induced fever, diminished SCV viral levels and reduced acute diffuse alveoli damage. The 10-40 mg/kg accumulated dosages of siRNA did not show any sign of siRNA-induced toxicity. These results suggest that a clinical investigation is warranted and illustrate the prospects for siRNA to enable a massive reduction in development time for new targeted therapeutic agents.
Article
Individual microRNA sequences can suppress the production of hundreds of proteins. Reduction of protein levels in this way is often modest, however, and many such RNAs probably collectively fine-tune gene expression.
Article
In conclusion, Argonaute proteins comprise a highly conserved protein family that is involved in a variety of RNA silencing phenomena in a diverse set of organisms. Perhaps through RNAi-related pathways or possibly also through distinct mechanisms, Argonaute family members have a variety of biological roles. These include regulation of development and determination of stem-cell character. Through an ever-advancing understanding of the biochemistry and genetics of the Argonaute family, we may ultimately reveal the reach of these pathways into the basic biological mechanisms that underlie a variety of human diseases.
Article
Double-stranded RNA (dsRNA) is an important regulator of gene expression in many eukaryotes. It triggers different types of gene silencing that are collectively referred to as RNA silencing or RNA interference. A key step in known silencing pathways is the processing of dsRNAs into short RNA duplexes of characteristic size and structure. These short dsRNAs guide RNA silencing by specific and distinct mechanisms. Many components of the RNA silencing machinery still need to be identified and characterized, but a more complete understanding of the process is imminent.
Article
We have constructed four different RNA polymerase III (Pol III)-based expression vectors, containing H1 or U6 promoters from human and mouse, which enable the endogenous production of small RNA transcripts for gene silencing applications. In addition, to facilitate the selection of recombinant clones, we have further improved these vectors by constructing a stuffer of puromycin resistance gene (Puro(r)) between ClaI and HindIII sites, which makes the preparation of vectors easy for rapid and efficient cloning of targeting sequences. A comparative analysis of the silencing efficiency between shRNA, sense-RNA, antisense-RNA, and siRNA showed that both the shRNA and siRNA, but not the sense-RNA and antisense-RNA, dramatically inhibit the targeting gene firefly luciferase activity in mammalian cells. However, there were no significant differences in the inhibition of firefly luciferase expression by shRNA and siRNA expressed from these DNA vectors. In summary, these improved DNA vector-based RNAi systems should provide a simple, convenient, and efficient cloning strategy for studying gene functions in mammalian cells.
Article
SiRNAs exert their biological effect by guiding the degradation of their cognate mRNA sequence, thereby shutting down the corresponding protein production (gene silencing by RNA interference or RNAi). Due to this property, siRNAs are emerging as promising therapeutic agents for the treatment of inherited and acquired diseases, as well as research tools for the elucidation of gene function in both health and disease. Because of their lethality and prevalence, lung diseases have attracted particular attention as targets of siRNA-mediated cures. In addition, lung is accessible to therapeutic agents via multiple routes, e.g., through the nose and the mouth, thus obviating the need for targeting and making it an appealing target for RNAi-based therapeutic strategies. The clinical success of siRNA-mediated interventions critically depends upon the safety and efficacy of the delivery methods and agents. Delivery of siRNAs relevant to lung diseases has been attempted through multiple routes and using various carriers in animal models. This review focuses on the recent progress in non-viral delivery of siRNAs for the treatment of lung diseases, particularly infectious diseases. The rapid progress will put siRNA-based therapeutics on fast track to the clinic.