Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

REVIEW ARTICLE

Engineered proteins with Pumilio/fem-3 mRNA binding


factor scaffold to manipulate RNA metabolism
Yang Wang1, Zefeng Wang1 and Traci M. Tanaka Hall2
1 Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC, USA
2 Laboratory of Structural Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park,
NC, USA

Keywords Pumilio/fem-3 mRNA binding factor proteins are characterized by a


protein engineering; protein–RNA sequence-specific RNA-binding domain. This unique single-stranded RNA
interaction; PUF protein; RNA; RNA
recognition module, whose sequence specificity can be reprogrammed, has
localization; RNA regulation; RNA-binding
been fused with functional modules to engineer protein factors with various
protein; splicing; translation; X-ray
crystallography functions. We summarize the advances made with respect to developing
RNA regulatory tools, as well as opportunities for the future.
Correspondence
Z. Wang, Department of Pharmacology,
Lineberger Comprehensive Cancer Center,
University of North Carolina at Chapel Hill,
Chapel Hill, NC, USA
Fax: +1 919 966 5640
Tel: +1 919 966 0131
E-mail: zefeng@med.unc.edu
or
T. M. Tanaka Hall, Laboratory of Structural
Biology, National Institute of Environmental
Health Sciences, National Institutes of
Health, Research Triangle Park, NC, USA
Fax: +1 301 480 3055
Tel: +1 919 541 1017
E-mail: hall4@niehs.nih.gov

(Received 28 January 2013, revised 29 May


2013, accepted 30 May 2013)

doi:10.1111/febs.12367

Introduction
Isaac Newton based his first Rule of Reasoning in Phi- modular molecule. In many key gene expression steps,
losophy on the teleological principle of purposeful such as DNA transcription or RNA splicing, the regu-
design and assertion that ‘Nature is pleased with sim- latory proteins usually comprise a nucleic acid binding
plicity’. Such simplicity is often illustrated in Nature’s module to recognize a target and a functional module
elegant solution to control gene expression with a to activate or inhibit the biochemical process. This sim-

Abbreviations
AAV, adeno-associated virus; ASRE, artificial site-specific RNA endonuclease; ESF, engineered splicing factor; FBF, fem-3 mRNA binding
factor; GFP, green fluorescent protein; miRNA, microRNA; PPR, pentatricopeptide repeat; PUF, Pumilio/FBF; RRM, RNA recognition motif;
RS, arginine/serine.

FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA. 3755
Designed PUF proteins Y. Wang et al.

ple design principle has also been adopted by humans the eight RNA-recognition helices recognizes one base
to engineer protein factors to manipulate DNA/RNA. using amino acid side chains at specific positions in
For specific recognition of RNA, Nature has evolved the second of three a-helices in each PUF repeat
an RNA-binding module, the Pumilio/FBF (PUF) (Fig. 1B) [22]. Two of these side chains interact specifi-
scaffold, whose specificity, itself both simple and modu- cally with an edge of the base, whereas a third side
lar, is determined by combination of functional base chain forms a stacking interaction with the base. The
recognition repeats. This unique RNA-binding mode second helix in each PUF repeat includes a five-residue
allows reprogramming of the specificity of the PUF sequence, designated as 12xx5, where the side chain at
scaffold with a small number of mutations, thus gener- the second position stacks with the recognized base
ating novel engineered protein factors to modulate and the side chains at the first and fifth positions
RNA metabolism. In this review, we present the design contact the edge of the RNA base [22,23].
principles, current progress and future outlook of engi- Mutagenesis confirmed that Nature has provided a
neering protein factors with a PUF domain as the remarkably simple code for base recognition: serine
RNA-binding scaffold. A broader review of many dif- and glutamate at the first and fifth positions recognize
ferent RNA-binding modules and their utility for engi- guanine; cysteine/serine and glutamine recognize ade-
neering specificity accompanies our more focused nine; and asparagine and glutamine recognize uracil
report [1]. (Fig. 1C) [22,24]. No code for cytosine was apparent
Drosophila melanogaster Pumilio and Caenorhabid- from Nature, although one of the repeats (repeat 4)
itis elegans FBF are the founding members of the can accept any base. In vitro binding affinity is tight as
PUF family of RNA regulatory proteins. Pumilio is a result of the many stacking interactions between pro-
encoded by a maternal gene required for abdominal tein side chains and RNA bases. Wild-type human
development in Drosophila [2]. FBF was selected by Pumilio1 binds to cognate RNA with a Kd of 0.5 nM,
yeast 3-hybrid as a protein that binds to an RNA whereas designed Pumilio1 mutants bind to their
sequence in the 3′ UTR of fem-3 mRNA [3,4], regulat-
ing the switch from spermatogenesis to oogenesis in
C. elegans hermaphrodites. Molecular cloning revealed
A B
that Pumilio and FBF proteins share a conserved
RNA-binding domain with eight sequence repeats of
approximately 36 amino acids, which was named a
Pumilio homology domain or PUF domain [4–7]. The
known target RNAs suggested that both proteins
recognize a sequence in the 3′ UTR containing a
conserved UGUR sequence (where R is a purine)
[4,6,8–11]. Pumilio and FBF regulate the stability or
translation of target mRNAs by recruiting effector
C
protein complexes to their target RNAs. The PUF
proteins themselves appear to lack additional func-
tional modules, and, instead, protein–protein interac-
tions, often with the RNA-binding domain, assemble
different complexes on the target RNAs [12–16]. The
formation of complexes may also fine tune RNA speci-
ficity [17]. PUF proteins utilize these activities to regu-
Fig. 1. PUF domain RNA interaction scaffold. (A) Ribbon diagram
late stem cell maintenance, cell proliferation and of a crystal structure of human Pumilio 1 RNA-binding domain in
differentiation, and the stress response [18,19]. complex with RNA ligand (5′-UGUACAUA-3′). RNA interaction
helices are shown as light blue cylinders and are labelled R1–R8.
Edge-interacting side chains from each repeat are coloured light
RNA recognition code for native PUF blue, stacking side chains are coloured pink, and RNA is coloured
proteins peach. For stick representations, nitrogen atoms are blue, oxygen
atoms are red, and sulfur atoms are yellow. (B) Interaction of PUF
The first crystal structures of PUF proteins revealed
repeats with uracil or adenine bases. Hydrogen bonds or van der
how the repeated sequences form a series of a-helical Waals contacts are indicated by dotted lines. (C) PUF RNA
repeats that assemble an arc of RNA-recognition heli- interaction code. Edge-interacting side chains that specify base
ces (Fig. 1A) [20,21]. A structure of human Pumilio1 recognition are shown. G, A and U were derived from natural PUF
in complex with an RNA ligand showed how each of proteins, whereas C recognition was selected by screening.

3756 FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA.
Y. Wang et al. Designed PUF proteins

cognate RNAs with a Kd in the range from 4 pM to Additional studies of the RNA target specificities of
18 nM. The effect on binding of a single noncognate different PUF proteins have revealed how Nature
repeat is largest when a base other than G is presented expands recognition properties using an assortment of
opposite a G-specific repeat (30–150-fold weaker) and variations on the theme of the 1 PUF repeat : 1 RNA
smallest when a G is presented opposite an A-specific base recognition observed for Pumilio 1 [26–28,30–35].
repeat (1.5–3-fold weaker) [24]. Other noncognate mis- Crystal structures of PUF proteins with RNA ligands
matches reduce binding by 10- to 20-fold [24,25], indi- demonstrate that extra or other nonspecifically recog-
cating the importance of most repeats for specificity. nized bases within natural PUF target sequences may
With this RNA recognition code, in principle, the stack directly with adjacent bases and/or be flipped
RNA-binding specificity of human Pumilio1 can be away from the PUF protein recognition surface
manipulated by site-directed mutagenesis to recognize [23,29,30,36,37]. A specialized binding pocket in a sub-
various RNA targets containing guanine, adenine and set of PUF proteins recognizes a cytosine base
uracil. upstream of the core sequence [30,38], and forming a
The identification of this recognition code and the complex with a partner protein can modify the recog-
ability to modify specificity of a PUF repeat has facili- nition motif [17]. The combinations of such variations
tated the identification of the RNA recognition proper- produce the unique specificity of natural target recog-
ties of other PUF proteins. PUF proteins comprise a nition by PUF proteins.
relatively small family of RNA-binding proteins with The accumulated body of evidence from genome-
few members in any organism. Humans and other wide RNA target identification, X-ray crystal struc-
mammals have two, whereas Drosophila has one. Sac- tures, in vitro RNA-binding assays and systematic
charomyces cerevisiae with six and C. elegans with nine mutagenesis studies indicate that PUF protein repeats
comprise larger families. This is in contrast to RNA rec- toward the N- and C-termini recognize conserved
ognition motif (RRM) proteins or hnRNP K homology RNA sequence elements, whereas plasticity in RNA
domains, where hundreds of proteins are found in an recognition is focused mainly at the central repeats 4
organism. Most PUF proteins are predicted to have and 5. The conserved 5′ UGUR sequence in PUF pro-
eight PUF repeats as in human Pumilio1, and the base tein target sequences is recognized by repeats 5–8
recognition side chains are highly conserved. However, (Fig. 1A). A downstream AU sequence is also con-
the RNA target sequences of these proteins are consid- served and recognized by repeats 2 and 3 in many
erably more diverse than would be expected from the PUF proteins. Valley et al. [34] examined systemati-
strong conservation. The core target sequences begin cally the effect of alanine mutation of either
with a 5′ UGUR but may contain from eight to 10 edge-interacting or base-stacking side chains on RNA-
bases with differing 3′ sequences. binding by C. elegans FBF and yeast Puf3p and
To reconcile this inconsistency, the Wickens labora- Puf4p. They found that repeats 4 and 5 can often tol-
tory at the University of Wisconsin used the design- erate alanine substitutions and proposed a ‘two-
ability of PUF repeat base recognition to determine handed model’ to describe PUF–RNA interactions,
the mode of recognition for additional C. elegans PUF where the N- and C-terminal PUF repeats ‘grasp’ the
proteins [26,27]. Beginning with FBF, the Wickens lab- conserved 3′ and 5′ sequences of RNA binding sites,
oratory used the yeast three-hybrid assay to measure respectively. Protein-specific variations in RNA
RNA binding of cleverly designed PUF proteins. They recognition are accommodated between them.
had determined that FBF with eight PUF repeats In the case of yeast Puf4p, plasticity in recognition
bound to a core RNA sequence with nine bases [28], allows binding to target RNAs with two different rec-
although the question remained as to whether the ognition patterns. In many target sites, the seventh of
‘extra’ base was positioned within the sequence or at nine bases is flipped away from the RNA-binding sur-
the 3′ end? To answer this question, they created face (seven-flipped), driven by recognition of an adeno-
designed FBF proteins where a specific repeat was sine at position 6 by repeat 3 (5′-UGUAUA-n7-UA,
mutated to recognize a different base, and the where n7 is any base at position 7). In a second set of
sequence specificity of the designed proteins was deter- target sites, the recognition pattern produces flipping
mined [27]. The change in sequence specificity versus of the sixth base (six-flipped) with an adenosine at
wild-type FBF established the register of PUF repeats position 7 recognized by repeat 3 (5′-UGUAU-n6-
and RNA bases. It was concluded that FBF targets AUA). The sequence of these six-flipped sites
possess an ‘extra’ base at the sixth of nine bases that is also matches the target sites of another yeast PUF
not recognized by the protein, and crystal structures protein, Mpt5p. Thus, Puf4p and Mpt5p may dually
have confirmed this [29,30]. regulate target mRNAs with six-flipped sites, whereas

FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA. 3757
Designed PUF proteins Y. Wang et al.

seven-flipped sites are not recognized by Mpt5p and PUF variants that specify cytosine binding. Each vari-
regulated only by Puf4p. ant pairs an arginine at the fifth position with a small
Stacking interactions in PUF-RNA recognition con- side chain at the first position. The ‘C code’ could be
tribute substantially to the binding energy, although transferred to different PUF repeats, and tyrosine, his-
the role of these interactions in binding specificity of tidine or arginine side chains at the second position of
PUF proteins is less well understood. RNA sequence the RNA interaction sequence are capable of forming
specificity can be changed by site-directed mutagenesis stacking interactions for cytosine recognition [25]. The
of only the two edge-interacting side chains [24]. How- identification of the C code makes it possible to design
ever, it appears that the identity of the third amino PUF domains that bind any eight-base RNA target
acid side chain that stacks with the bases has been sequence and increases the potential for use of engi-
optimized for the natural specificities of PUF proteins neered PUF proteins in research and therapeutics.
[39]. Substitution of the stacking side chains in C. ele-
gans FBF broadens the range of RNA bases that can
Engineered factors with PUF as
be tolerated at a particular position. The simple
RNA-binding scaffold
approach of designing PUF specificity by altering
edge-interacting side chains is typically successful, The unique RNA-binding mode of the PUF domain
although a better understanding of how stacking side makes it possible to engineer artificial proteins to mod-
chains contribute to specificity may enhance the target ulate RNA metabolism. Following the composition of
selectivity of designed proteins. natural proteins, these artificial proteins employ a
modular design by fusing a functional module (protein
domains with desired activities) to a target recognition
Expanding the modular binding code
module (modified PUF domains with re-designed
for cytosine
RNA binding specificity). Compared with other
With the natural modularity and recognition code of domains that specifically bind single-stranded RNA,
human Pumilio 1, its PUF domain can be engineered such as RRM, Zn finger and pentatricopeptide repeats
to target an eight-nucleotide RNA sequence containing (PPR), the PUF domain has the advantage of having
adenine, uracil and guanine [22,24]. By fusing the PUF a fully determined modular binding code. The RRM
domain with a functional domain, these ‘designer’ and Zn finger domains only recognize short RNA
PUFs have been applied to track RNA localization in motifs (3–4 nucleotides), and their binding specificity
cells [40,41], regulate alternative splicing [25,42] and cannot be reprogrammed. However, it is conceivable
modulate translational regulation [43]. Opportunities that several RRMs or Zn fingers can be tandemly
remain to develop novel tools for biomedical research linked to recognize a longer RNA. Similar to the PUF
with potential therapeutic applications (see below). domain, the PPR domain can recognize RNA in a
A limitation to the application of designer PUFs modular fashion and several studies have begun to
had been that no recognition code for cytosine speci- make progress with respect to solving its modular
ficity was found in Nature. However, two groups binding code [45]. Once the binding code is completed,
recently succeeded in identifying combinations of the PPR may be used similarly to PUF as a modular
amino acid side chains that specify cytosine recogni- RNA binding scaffold in engineered factors. The main
tion by a PUF repeat, expanding the PUF recognition disadvantage of PUF is that the native protein
code and permitting the creation of PUFs directed to contains only eight repeats that recognize 8-nucleotide
any eight-base RNA sequence [25,44]. Both groups target; however, it is possible to expand the length of
used yeast three-hybrid screening of a PUF domain PUF recognition sites (described below). Varying the
library with random sequences at the first and fifth choice of functional modules, an assortment of PUF
positions of the RNA interaction motif in repeat 6 to factors has been engineered since 2007 and more fac-
select proteins that bound a cytosine at the corre- tors with novel activities are expected to be generated
sponding third base of the RNA target. Dong et al. in the future.
[25] found that a combination of serine at the first
position and arginine at the fifth position specify cyto-
Engineered PUF proteins as specific RNA probes
sine. A crystal structure showed hydrogen bonding
between the side chain of arginine and the O2 and N3 The first application of artificial PUF proteins was as
positions of the cytosine. The serine residue positions a fluorescent marker to visualize RNAs in live cells
the arginine but does not contact the RNA. Using a [40]. A highly effective approach to visualize RNAs in
similar strategy, Filipovska et al. [44] selected five live cells is to use a fusion protein of green fluorescent

3758 FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA.
Y. Wang et al. Designed PUF proteins

protein (GFP) fused to bacteriophage coat protein domains with split mCitrine and used it to reveal plant
MS2 that recognizes reporter RNAs containing multi- viral RNA localization. Another variation of this
ple copies (e.g. 24 copies) of MS2 hairpin sequences method used PUF domains fused with split luciferase
[46–48]. However, the GFP-MS2 fusion protein to detect ssRNA in vitro [51]; however, this approach
requires artificial RNA targets with MS2 hairpins. To has not yet been tested in live cells.
visualize endogenous RNA targets, Ozawa et al. [40] The use of split fluorescent protein reduces the noise
designed a split GFP system that comprises two fusion level of RNA detection because the fluorescent protein
proteins, each containing a PUF domain and either an is assembled by the RNA target. However, because of
N-terminal or C-terminal fragment of GFP (Fig. 2A). the limited efficiency in the co-folding of two GFP
When the PUF domains recognize the adjacent fragments and only one GFP per RNA, this system
8-nucleotide sequences in the same RNA, the two produces a lower fluorescent signal compared to the
fusion proteins are brought together so that the split GFP-MS2. On the other hand, the PUF-split GFP can
GFP fragments assemble and fluoresce. The specificity detect a single endogenous RNA target, whereas the
of this system is increased by requiring binding to two GFP-MS2 requires a reporter RNA with multiple
8-nucleotide sites separated by a small spacer. This MS2 target sites to produce a higher signal over the
design has been used to visualize mitochondrial RNA diffuse background noise of unbound fluorescent pro-
in cultured mammalian cells [40] and cytosolic b-actin tein. For some special cases, it should be possible to
mRNA [49], and a modified version using intact GFP use the GFP-PUF fusion protein directly. Similar to
fused with two PUFs was also used to detect cytosolic the GFP-MS2 fusion protein, the GFP-PUF will pro-
b-actin mRNA [50]. A similar approach was also duce a weak fluorescence background. However, if the
developed by Tilsner et al. [41] who combined PUF target contains multiple copies of the recognition site,

Fig. 2. Engineered PUF factors. (A)


Fluorescent probe for in vivo RNA
labelling. Combination of split GFP (or
other fluorescent protein) with PUF
scaffold generated an RNA probe to
visualize RNA in live cells. (B) Engineered
splicing factors. Combination of a PUF C
scaffold with an RS domain or a Gly-rich
domain generated splicing factors that
activate or inhibit splicing of alternative
exon. (C) Modulation of translation. Fusion
of GLD2 or CAF1 with PUF domain
produced novel factors that can activate or
inhibit mRNA translation. (D) Artificial site- D
specific RNA endonuclease. Combination
of PUF domain with a nonspecific RNA
endonuclease (PIN domain) can produce a
new class of enzymes that specifically
recognize and cleave RNA.

FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA. 3759
Designed PUF proteins Y. Wang et al.

the GFP-PUF may form bright foci on the target Another elegant use of the modular configuration
RNA, as with the GFP-MS2 protein that forms a for RNA regulation was developed by Cooke et al.
bright spot on reporter RNAs with consecutive MS2 [43] to alter translation. They attached a PUF domain
hairpins [47]. Several trinucleotide expansions have to either a translational activator, GLD2, or a transla-
been found to cause neurodegenerative diseases (such tional repressor, CAF1 (Fig. 2C). The resulting PUF-
as CAG repeats in Huntington disease and CUG GLD2 engineered proteins specifically recognized their
repeats in myotonic dystrophy) [52]. These pathogenic RNA target and activated translation and induced
RNA repeats usually contain hundreds of copies of polyadenylation in Xenopus oocytes, whereas the PUF-
the trinucleotide; thus, a GFP-PUF recognizing these CAF1 fusion protein repressed translation and directed
repeated sequences may provide a new way to visualize deadenylation. The coordinate regulation of transla-
the RNA and study the dynamics of such pathogenic tion and/or stability of mRNA by PUF proteins and
RNA in live cells. microRNA (miRNAs) [54–57] suggests that engineered
PUF proteins may be designed to use as antagonizers
or enhancers of miRNA regulation.
Engineered PUF proteins as regulatory factors for
gene expression
Engineered PUF proteins as novel RNA
Mammalian gene expression is controlled by multiple
endonucleases
regulatory steps during mRNA processing by protein
factors with modular configuration: separable RNA Specific cleavage of RNAs is critical for in vitro
recognition and functional domains. A common manipulation of RNA and for in vivo gene silencing.
method for studying the function of RNA regulatory However, a simple enzyme that cleaves RNA in a
proteins is to tether the protein or a fragment to a spe- sequence-specific manner has not been found in Nature,
cific RNA target and analyze the consequences. Using despite extensive investigations. Most RNA endonuc-
this engineering principle, artificial regulatory factors leases either have limited sequence specificity (e.g.
can be derived by combining a PUF domain with dif- RNase A or RNase T1) [58–60] or recognize their tar-
ferent effector domains. For example, Wang et al. [42] gets by specific structures (e.g. tRNA splicing endonu-
developed engineered splicing factors (ESFs) by fusing clease) [61] or through guide RNA that pairs with target
a PUF domain with Arg/Ser-rich (RS) domains of (e.g. Argonaute proteins) [62]. By fusing a PUF domain
SRSF1 or the glycine-rich (Gly) domain of hnRNP A1 with a nonspecific endoribonuclease domain (PIN
(Fig. 2B). The resulting ESFs can function either as domain of Smg6p), Choudhury et al. [63] engineered
splicing activators (PUF-RS) or inhibitors (PUF-Gly), artificial site-specific RNA endonucleases (ASREs) that
and can specifically control different types of alterna- specifically recognize RNA substrates and efficiently
tive splicing. Specifically, PUF-RS promotes exon cleave near the binding sites both in vitro and in cul-
inclusion when binding to an alternative exon but tured cells (Fig. 2D) [63]. The digested products have
inhibits splicing when binding downstream of an alter- 5′-phosphate and 3′-hydroxyl groups, making it possible
native exon. PUF-Gly, on the other hand, inhibits to religate. Two ASREs were designed to silence specifi-
exon inclusion when binding to the target RNA either cally the expression of a bacterial gene or human mito-
inside or downstream of the alternative exon [42,53]. chondrial mRNAs that contain one or two binding sites
When binding to the region between two alternative of designer PUFs. Because PUF domains recognize
5′ or 3′ splice sites, PUF-RS promotes use of the prox- their targets through an 8-nucleotide sequence, compa-
imal splice site to favour the longer splicing isoform, rable in length to the seed match of small interfering
whereas PUF-Gly inhibits proximal site usage to RNA, engineered ASREs may serve as an RNA silenc-
induce the shorter isoform [42]. ing tool complementary to RNA interference, which will
Using this general approach, a PUF-Gly ESF was be effective in organisms or cellular compartments
generated to shift splicing of the Bcl-x pre-mRNA from where RNA interference machinery is not present.
the anti-apoptotic long isoform (Bcl-xL) to the pro-
apoptotic short isoform (Bcl-xS). The altered splice
Future application of PUF-based
isoform distribution was sufficient to sensitize several
designer factors
cancer cell lines to multiple anti-cancer drugs [42]. With
the identification of the C code, both PUF-RS and Mammalian gene expression contains multiple RNA
PUF-Gly ESFs were engineered to shift splicing of vas- processing stages, including RNA splicing, polyadeny-
cular endothelial growth factor-A pre-mRNA from the lation, editing, translocation, translation and degrada-
angiogenic to anti-angiogenic isoform [25]. tion, which are kept under tight control by a variety of

3760 FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA.
Y. Wang et al. Designed PUF proteins

RNA-binding proteins. The reprogrammable RNA- process is tightly regulated, and the mis-regulation of
binding scaffold of the PUF domain makes it possible splicing is a common cause of human diseases [66].
(and tempting) to create artificial factors that can spe- The specific manipulation of alternative splicing will
cifically modulate target RNAs at each processing provide a useful way to fine tune gene function. Many
stage. A benefit of manipulating gene expression at the known splicing factors have modular activities in
RNA level rather than the DNA level is that the effect which they recognize a short RNA element in the tar-
is often nonpermanent and reversible. The factors can get with an RNA-binding module (e.g. RRM or
be engineered by fusing designer PUF domains with a hnRNP K homology domain) and alter splicing with a
protein factor (or its functional domain) with known functional module. The best-known functional
regulatory activity on RNA metabolism. In theory, any domains for splicing modulation are the RS domains
protein that has demonstrated activity in a tethering and Gly-rich domains, which were used to develop the
experiment can be fused with PUF domains to produce first generation of engineered splicing factors [25,42].
novel artificial factors that target endogenous RNA However, other domains in splicing regulatory factors
targets. Minimal functional domains can be determined may have distinct activities in regulating splicing. For
by fusing fragments of known regulatory factors to example, we found that the C-terminal domain of sev-
PUF domains and testing activity. Alternatively, new eral SR proteins can activate or inhibit splicing when
functional modules can be identified by constructing a binding to different pre-mRNA regions [53], the ala-
cDNA library of PUF domain fusions and screening nine-rich motif of RBM4 can inhibit splicing, and the
for RNA regulation. A previous review has proposed proline-rich motif of DAZAP1 can enhance splicing
several ideas for potential application of engineered [67]. These new functional domains can be used to
RNA-binding proteins [64], some of which have been derive additional types of artificial splicing factors to
achieved successfully using the PUF scaffold (Fig. 2). fine tune alterative splicing (Fig. 3B).
In addition, we propose other potential applications
using the engineered PUF factors in humans, and simi-
Engineered factors to control alternative
lar applications can be adapted to other organisms.
polyadenylation
Many human genes contain multiple polyadenylation
Engineering RNA editing enzymes
sites. The use of different polyadenylation sites creates
RNA editing is commonly found in many eukaryotes to mRNA isoforms with different 3′ UTRs that regulate
generate specific sequence substitutions and changes in the translation efficiency and stability of mRNA. Simi-
gene expression levels, resulting in an increase of RNA lar to alternative splicing, control of alternative poly-
and protein diversity. Adenosine-to-inosine (A-to-I) adenylation can have multifaceted effects on biological
editing represents the most important class of RNA processes, and shortening of the 3′ UTR through prox-
editing in human and affects function of many genes, imal polyadenylation site usage is closely regulated in
especially in the central nervous system [65]. The native cell proliferation and differentiation [68–70]. Thus
substrate for A-to-I editing is a double-stranded RNA reprogramming the 3′ UTR region by modulating
region, recognized by the adenosine deaminase acting polyadenylation site selection may provide another
on RNA. Canonical adenosine deaminase acting on way to manipulate gene expression. Several recent
RNA proteins have a conserved catalytic deaminase studies suggested that alternative polyadenylation is
domain and one or more double-stranded RNA-binding regulated by trans-acting factors that recognize cis-
domains; however, it has not been determined whether elements near polyadenylation sites [71–73]. Some of
these domains function in a modular fashion [65]. Thus, these factors can control both splicing and poly-adeny-
we propose replacing the double-stranded RNA-binding lation (eg. U1A, PTB) [71,73]. Therefore, we expect
domains with a PUF domain and testing whether the engineered PUF fusion proteins to be similarly success-
resulting fusion factors have deaminase activities against ful in modulating the use of alternative polyadenyla-
single-stranded RNA (Fig. 3A). With optimization, the tion sites (Fig. 3C).
engineered enzyme may specifically recognize an RNA
target by sequence and deaminate a nearby adenosine.
Engineered factors to control RNA localization
The transport of RNA to correct intracellular loca-
Next-generation engineered splicing factors
tions is an essential step for the expression of both
Most human genes undergo alternative splicing to pro- coding and noncoding messages because the function
duce multiple isoforms with distinct activities. This of RNA is highly localized. Although there are many

FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA. 3761
Designed PUF proteins Y. Wang et al.

B Fig. 3. Future engineered factors with


PUF scaffold as RNA recognition module.
(A) Novel enzyme for single-stranded RNA
editing. Sequence specificity may be
determined by a PUF binding site rather
than double-stranded RNA. (B) Next-
generation engineered splicing factors.
C With recently identified functional domains
that control splicing in diverse fashion,
new splicing factors may be generated to
fine tune alternative splicing. (C)
Engineered PUF factors to manipulate
alternative polyadenylation. By combining
a PUF domain with regulatory proteins/
D domains for alternative polyadenylation
sites, artificial factors may be constructed
to inhibit or activate certain
polyadenylation sites and thus change the
3′ UTR of mRNA. (D) Engineered RNA
transporters. New proteins can be
engineered by combining a PUF domain
with protein translocation signals to
E transport RNA into different cellular
compartments. (E) Engineered PUF factors
to control RNA degradation. The PUF
domain can be linked with additional
domains to specifically recruit RNA
stabilization or destabilization proteins,
F thus controlling RNA half-life. (F)
Engineered PUF factors to control
transcription elongation. By specifically
recognizing the nascent RNA transcript,
new PUF factors can be engineered to
induce transcription pausing or release
paused RNA polymerases.

known RNA elements that govern their transport and nuclear localization sequence may be created to block
final localization, other recognition elements are nuclear-to-cytoplasmic RNA transport. Such engi-
unknown, although the detailed mechanisms are neered proteins can be designed to recognize the loop-
unclear. On the other hand, protein localization signals region of a specific pre-miRNA, thus retaining
are better understood and well characterized signal the pre-miRNA in the nucleus and preventing its
peptides or fragments have been established that are processing to mature form in the cytoplasm, thereby
responsible for protein localization in nuclear or cyto- inhibiting the miRNA function.
plasmic compartments or for protein secretion. There-
fore, by combining these established protein
Engineered PUF factors to affect RNA stability
localization signals with PUF domains, we may create
engineered factors to specifically transport RNA Turnover of mRNA is continuous in the cytoplasm
(Fig. 3D). For example, a PUF domain fused with a and often regulated by protein factors that recognize

3762 FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA.
Y. Wang et al. Designed PUF proteins

the 3′ UTR region. A well-studied case is directed by domains and split GFP fragments [40] and was used
AU-rich elements in the 3′ UTR of short-lived mRNA, subsequently by creating a fusion protein containing
which destabilize the messages [74]. Several factors two PUF domains separated by an intact GFP protein
positively or negatively regulate this process, and PUF [50]. Specificity for a 16-nucleotide sequence should
fusion proteins with fragments of these factors may ensure the recognition of a unique target in the human
function as engineered factors to modulate RNA transcriptome. However, engineered factors with tan-
stability (Fig. 3E). dem PUF domains may recognize an 8-nucleotide
‘half-site’ with sufficient affinity for functional activity.
An alternative approach is to construct an expanded
Engineered PUF transcriptional regulators
PUF domain by inserting more PUF repeats into a
RNA transcription can be controlled at the initiation- native PUF domain. For example, a PUF domain with
to-elongation switch of RNA polymerase II through a 16 repeats was demonstrated to bind cognate RNA
transcriptional pausing step after the initiation and target in a yeast-three-hybrid assay [44], and it should
synthesis of a small RNA fragment [75]. Such pausing be possible to construct PUF domains with different
is released to continue transcription, which is often numbers of repeats using similar design. We have gen-
controlled by transcription elongation factors. Using erated expanded PUF domains with 9, 10 or 12 PUF
PUF engineered factors that recognize the nascent repeats and found that these novel PUF scaffolds
transcripts, it may be possible to design factors that can indeed recognize cognate targets (W. Zhang and
specifically control transcription at the initiation-to- Z. Wang, unpublished results).
elongation switch by promoting or releasing the Although most applications will benefit from a PUF
paused RNA polymerase II (Fig. 3F). For example, domain with increased specificity, in certain cases, the
the release of paused RNA polymerase II can be medi- engineered PUF factors may need to recognize multi-
ated by CDK9 and cyclin T1, which form a tightly ple diverse targets with a degenerate binding code (i.e.
complex to promote transcription elongation [76]. It a decreased specificity). This requires generating PUF
may be possible to recruit CDK9 through a PUF- domains that recognize shorter target sequences. Two
CDK9 fusion protein to the nascent transcripts, which strategies may solve this problem. One aims to identify
in turn releases paused polymerase II and activates amino acid side chain combinations in PUF repeats
transcription elongation. that can tolerate multiple or all bases to produce a
binding code for ‘N’. This degenerate code can be
used in combination with the specific base recognition
Additional challenges for engineered
code to make new PUF domains that specifically rec-
PUF factors
ognize sequences shorter than 8-nucleotides at the
same time as maintaining binding affinity. We suggest
Expanding binding specificity
that it may be possible to screen for a degenerate code
Most naturally occurring PUF proteins have eight of PUF-RNA interaction. Another approach is to
RNA-binding repeats, which recognize eight bases in construct a PUF scaffold with fewer than eight PUF
target mRNA. Some PUF domains can accommodate repeats. The minimum number of repeats in a stable
one or two extra bases; however, the specificity of the PUF scaffold is unknown, although we speculate that
extra bases cannot be reprogrammed. Thus, the a minimal number may be required for correct fold-
sequence specificity of engineered PUF factors is lar- ing. Using a PUF scaffold with fewer repeats also
gely determined by an 8-nucleotide PUF binding site, decreases the size of the engineered protein, which
which is comparable to the specificity of miRNA that may be beneficial for in vivo application and simplify
recognize targets by 7-nucleotide seed match. How- the design.
ever, similar to miRNA, designer PUF factors may
have off-target effects because, on average, any given
Minimizing off-target effects
8-nucleotide sequence will occur by chance in
sequences with 48 bases or 64 kb in length. The control of off-target effects is an important con-
Nevertheless, it is possible to increase the specificity cern for any method that specifically targets endoge-
of engineered PUF factors. One obvious approach is nous gene expression, especially when the engineered
to use two PUF domains in combination, which, in PUF factors are used in vivo. In addition to using a
theory, provides a recognition site of 16 nucleotides. PUF scaffold with higher specificity (e.g. a scaffold
This design was used first for the visualization of containing additional repeats), careful experimental
RNA in vivo with two fusion proteins of PUF design should minimize any off-target effects.

FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA. 3763
Designed PUF proteins Y. Wang et al.

The first opportunity to minimize off-target effects is Several nonviral methods may be used to deliver
within the design stage by identifying a target sequence DNAs or proteins into animals, and some of these
with fewer off-target hits in unrelated RNAs. Because methods are being tested for use in humans. These
the sequence of the human genome is not random, methods include different nano-particles and cell pene-
some sequence patterns are more common than others. tration peptides, and more in depth information on
When selecting PUF recognition sites in RNA targets, in vivo delivery methods is provided elsewhere [79–81].
it will be helpful to search the transcriptome and choose These methods can be applied, at least in principle, for
relatively uncommon sites. Although off-target effects in vivo delivery of the engineered PUF factors. Because
are impossible to eliminate, such a search will estimate the application of engineered PUF factors is still in its
the frequency of possible off-target sites. Another con- early stages, we expect the technology for protein
cern with respect to selecting target sequences is evalu- delivery to advance ahead of the in vivo application of
ating how RNA structure may affect PUF binding. these factors in live animals or humans.
Although the PUF scaffold can recognize an RNA
target sequence in a double-stranded region [44,54]
Final comments
(Z. Wang, unpublished results), the occurrence of
extensive hairpin structures in the binding site greatly Thus far, engineered factors combining a PUF scaffold
decreases binding affinity (Z. Wang, unpublished with a functional module have proven to be versatile
results). Therefore, we should select target sites in less tools for manipulating and understanding RNA
structured regions, whereas possible off-target sites in metabolism. Recent advances allowing the design of
more structured regions may not pose a problem. the PUF scaffold to recognize any RNA sequence
Another method for reducing off-target effects is to expand the potential to apply this technology for regu-
control the expression of the engineered PUF factors lating RNA processing pathways. Starting with the
by expressing them at the same time and place as their solution found in Nature, continued adherence to the
targets. For example, by designing an ASRE with rule of simplicity may be prudent in moving this new
mitochondrial targeting signals to silence mitochon- field forward.
drial RNA, we produced a protein with low off-target
effects against nuclear or cytoplasmic RNA because
Acknowledgements
the ASRE is undetectable outside mitochondria [63].
A wide array of gene expression tools have been devel- This research was supported in part by the Intramural
oped over the years to fine tune the expression of Research Program of the NIH, National Institute of
exogenous proteins (such as cell type specific vectors). Environmental Health Sciences (TMTH).
They should be helpful tools to fine tune the expres-
sion pattern of the engineered PUF factors.
References
1 Chen Y & Varani G (2013) Engineering RNA-binding
In vivo delivery of engineered PUF factors
proteins for biology. FEBS J doi:10.1111/febs.12375.
Because engineered PUF factors are typical proteins, 2 Lehmann R & Nusslein-Volhard C (1991) The maternal
they should be deliverable to live animals using estab- gene nanos has a central role in posterior pattern
lished gene therapy vectors. For application in cul- formation of the Drosophila embryo. Development 112,
tured cells or animal models, the lentiviral system is a 679–691.
good choice for ease of use and robust expression in 3 Ahringer J & Kimble J (1991) Control of the sperm-
most cell types. For therapeutic application in humans, oocyte switch in Caenorhabditis elegans hermaphrodites
by the fem-3 3′ untranslated region. Nature 349,
adeno-associated virus (AAV) is a favourable choice in
346–348.
gene therapy because it lacks known pathogenicity,
4 Zhang B, Gallegos M, Puoti A, Durkin E, Fields S,
can infect nondividing cells, and has many serotypes
Kimble J & Wickens MP (1997) A conserved RNA-
that allow specific gene delivery into different tissues.
binding protein that regulates sexual fates in the
The AAV can stably integrate into the host cell gen-
C. elegans hermaphrodite germ line. Nature 390, 477–484.
ome at a specific site (designated AAVS1) in human 5 Macdonald PM (1992) The Drosophila pumilio gene: an
chromosome 19 [77,78]; however, the non-integrating unusually long transcription unit and an unusual
AAV vectors are used for all gene therapy applica- protein. Development 114, 221–232.
tions. The drawback of AAV is its small package 6 Zamore PD, Williamson JR & Lehmann R (1997) The
capacity, although the currently engineered PUF Pumilio protein binds RNA through a conserved
factors are sufficiently small to be packed by AAV.

3764 FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA.
Y. Wang et al. Designed PUF proteins

domain that defines a new class of RNA-binding 22 Wang X, McLachlan J, Zamore PD & Hall TMT
proteins. RNA 3, 1421–1433. (2002) Modular recognition of RNA by a human
7 Barker DD, Wang C, Moore J, Dickinson LK & pumilio-homology domain. Cell 110, 501–512.
Lehmann R (1992) Pumilio is essential for function but 23 Lu G & Hall TMT (2011) Alternate modes of cognate
not for distribution of the Drosophila abdominal RNA recognition by human PUMILIO proteins.
determinant Nanos. Genes Dev 6, 2312–2326. Structure 19, 361–367.
8 Zamore PD, Bartel DP, Lehmann R & Williamson JR 24 Cheong CG & Hall TMT (2006) Engineering RNA
(1999) The PUMILIO-RNA interaction: a single RNA- sequence specificity of Pumilio repeats. Proc Natl Acad
binding domain monomer recognizes a bipartite target Sci USA 103, 13635–13639.
sequence. Biochemistry 38, 596–604. 25 Dong S, Wang Y, Cassidy-Amstutz C, Lu G, Bigler R,
9 Murata Y & Wharton RP (1995) Binding of pumilio to Jezyk MR, Li C, Hall TMT & Wang Z (2011) Specific
maternal hunchback mRNA is required for posterior and modular binding code for cytosine recognition in
patterning in Drosophila embryos. Cell 80, 747–756. Pumilio/FBF (PUF) RNA-binding domains. J Biol
10 Sonoda J & Wharton RP (1999) Recruitment of Chem 286, 26732–26742.
Nanos to hunchback mRNA by Pumilio. Genes Dev 13, 26 Koh YY, Opperman L, Stumpf C, Mandan A, Keles S
2704–2712. & Wickens M (2009) A single C. elegans PUF protein
11 Wharton RP, Sonoda J, Lee T, Patterson M & Murata binds RNA in multiple modes. RNA 15, 1090–1099.
Y (1998) The Pumilio RNA-binding domain is also a 27 Opperman L, Hook B, DeFino M, Bernstein DS &
translational regulator. Mol Cell 1, 863–872. Wickens M (2005) A single spacer nucleotide
12 Van Etten J, Schagat TL, Hrit J, Weidmann CA, determines the specificities of two mRNA regulatory
Brumbaugh J, Coon JJ & Goldstrohm AC (2012) proteins. Nat Struct Mol Biol 12, 945–951.
Human Pumilio proteins recruit multiple deadenylases 28 Bernstein D, Hook B, Hajarnavis A, Opperman L &
to efficiently repress messenger RNAs. J Biol Chem Wickens M (2005) Binding specificity and mRNA
287, 36370–36383. targets of a C. elegans PUF protein, FBF-1. RNA 11,
13 Blewett NH & Goldstrohm AC (2012) A eukaryotic 447–458.
translation initiation factor 4E-binding protein 29 Wang Y, Opperman L, Wickens M & Hall TMT (2009)
promotes mRNA decapping and is required for PUF Structural basis for specific recognition of multiple
repression. Mol Cell Biol 32, 4181–4194. mRNA targets by a PUF regulatory protein. Proc Natl
14 Weidmann CA & Goldstrohm AC (2012) Drosophila Acad Sci USA 106, 20186–20191.
Pumilio protein contains multiple autonomous 30 Qiu C, Kershner A, Wang Y, Holley CP, Wilinski
repression domains that regulate mRNAs independently D, Keles S, Kimble J, Wickens M & Hall TMT
of Nanos and brain tumor. Mol Cell Biol 32, 527–540. (2012) Divergence of Pumilio/fem-3 mRNA binding
15 Goldstrohm AC, Seay DJ, Hook BA & Wickens M factor (PUF) protein specificity through variations
(2007) PUF protein-mediated deadenylation is catalyzed in an RNA-binding pocket. J Biol Chem 287,
by Ccr4p. J Biol Chem 282, 109–114. 6949–6957.
16 Goldstrohm AC, Hook BA, Seay DJ & Wickens M 31 Gerber AP, Herschlag D & Brown PO (2004) Extensive
(2006) PUF proteins bind Pop2p to regulate messenger association of functionally and cytotopically related
RNAs. Nat Struct Mol Biol 13, 533–539. mRNAs with Puf family RNA-binding proteins in
17 Campbell ZT, Bhimsaria D, Valley CT, Rodriguez- yeast. PLoS Biol 2, E79.
Martinez JA, Menichelli E, Williamson JR, Ansari AZ 32 Kershner AM & Kimble J (2010) Genome-wide analysis
& Wickens M (2012) Cooperativity in RNA–protein of mRNA targets for Caenorhabditis elegans FBF, a
interactions: global analysis of RNA binding specificity. conserved stem cell regulator. Proc Natl Acad Sci USA
Cell Rep 1, 570–581. 107, 3936–3941.
18 Miller MA & Olivas WM (2011) Roles of Puf proteins 33 Stumpf CR, Kimble J & Wickens M (2008) A
in mRNA degradation and translation. Wiley Caenorhabditis elegans PUF protein family with distinct
Interdiscip Rev RNA 2, 471–492. RNA binding specificity. RNA 14, 1550–1557.
19 Wickens M, Bernstein DS, Kimble J & Parker R (2002) 34 Valley CT, Porter DF, Qiu C, Campbell ZT, Hall TMT
A PUF family portrait: 3′ UTR regulation as a way of & Wickens M (2012) Patterns and plasticity in RNA–
life. Trends Genet 18, 150–157. protein interactions enable recruitment of multiple
20 Wang X, Zamore PD & Hall TMT (2001) Crystal proteins through a single site. Proc Natl Acad Sci USA
structure of a Pumilio homology domain. Mol Cell 7, 109, 6054–6059.
855–865. 35 Yosefzon Y, Koh YY, Chritton JJ, Lande A, Leibovich
21 Edwards TA, Pyle SE, Wharton RP & Aggarwal AK L, Barziv L, Petzold C, Yakhini Z, Mandel-Gutfreund
(2001) Structure of Pumilio reveals similarity between Y, Wickens M et al. (2011) Divergent RNA binding
RNA and peptide binding motifs. Cell 105, 281–289. specificity of yeast Puf2p. RNA 17, 1479–1488.

FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA. 3765
Designed PUF proteins Y. Wang et al.

36 Gupta YK, Nair DT, Wharton RP & Aggarwal AK protein-tagged pumilio. ACS Chem Biol 7,
(2008) Structures of human Pumilio with noncognate 999–1005.
RNAs reveal molecular mechanisms for binding 51 Furman JL, Badran AH, Ajulo O, Porter JR, Stains
promiscuity. Structure 16, 549–557. CI, Segal DJ & Ghosh I (2010) Toward a general
37 Miller MT, Higgin JJ & Hall TMT (2008) Basis of approach for RNA-templated hierarchical assembly of
altered RNA-binding specificity by PUF proteins split-proteins. J Am Chem Soc 132, 11692–11701.
revealed by crystal structures of yeast Puf4p. Nat Struct 52 Galka-Marciniak P, Urbanek MO & Krzyzosiak WJ
Mol Biol 15, 397–402. (2012) Triplet repeats in transcripts: structural insights
38 Zhu D, Stumpf CR, Krahn JM, Wickens M & Hall into RNA toxicity. Biol Chem 393, 1299–1315.
TMT (2009) A 5′ cytosine binding pocket in Puf3p 53 Wang Y, Xiao X, Zhang J, Choudhury R, Robertson
specifies regulation of mitochondrial mRNAs. Proc A, Li K, Ma M, Burge CB & Wang Z (2013) A
Natl Acad Sci USA 106, 20192–20197. complex network of factors with overlapping affinities
39 Koh YY, Wang Y, Qiu C, Opperman L, Gross L, Hall represses splicing through intronic elements. Nat Struct
TMT & Wickens M (2011) Stacking interactions in Mol Biol 20, 36–45.
PUF-RNA complexes. RNA 17, 718–727. 54 Kedde M, van Kouwenhove M, Zwart W, Oude
40 Ozawa T, Natori Y, Sato M & Umezawa Y (2007) Vrielink JA, Elkon R & Agami R (2010) A Pumilio-
Imaging dynamics of endogenous mitochondrial RNA induced RNA structure switch in p27-3′ UTR controls
in single living cells. Nat Methods 4, 413–419. miR-221 and miR-222 accessibility. Nat Cell Biol 12,
41 Tilsner J, Linnik O, Christensen NM, Bell K, Roberts 1014–1020.
IM, Lacomme C & Oparka KJ (2009) Live-cell imaging 55 Leibovich L, Mandel-Gutfreund Y & Yakhini Z (2010)
of viral RNA genomes using a Pumilio-based reporter. A structural-based statistical approach suggests a
Plant J 57, 758–770. cooperative activity of PUM1 and miR-410 in human
42 Wang Y, Cheong CG, Hall TMT & Wang Z (2009) 3′-untranslated regions. Silence 1, 17.
Engineering splicing factors with designed specificities. 56 Miles WO, Tschop K, Herr A, Ji JY & Dyson NJ
Nat Methods 6, 825–830. (2012) Pumilio facilitates miRNA regulation of the
43 Cooke A, Prigge A, Opperman L & Wickens M (2011) E2F3 oncogene. Genes Dev 26, 356–368.
Targeted translational regulation using the PUF 57 Nolde MJ, Saka N, Reinert KL & Slack FJ (2007) The
protein family scaffold. Proc Natl Acad Sci USA 108, Caenorhabditis elegans pumilio homolog, puf-9, is
15870–15875. required for the 3′UTR-mediated repression of the let-7
44 Filipovska A, Razif MF, Nygard KK & Rackham O microRNA target gene, hbl-1. Dev Biol 305, 551–563.
(2011) A universal code for RNA recognition by PUF 58 Marshall GR, Feng JA & Kuster DJ (2008) Back to the
proteins. Nat Chem Biol 7, 425–427. future: ribonuclease A. Biopolymers 90, 259–277.
45 Barkan A, Rojas M, Fujii S, Yap A, Chong YS, Bond 59 Yoshida H (2001) The ribonuclease T1 family. Methods
CS & Small I (2012) A combinatorial amino acid code Enzymol 341, 28–41.
for RNA recognition by pentatricopeptide repeat 60 Beintema JJ & Kleineidam RG (1998) The ribonuclease
proteins. PLoS Genet 8, e1002910. A superfamily: general discussion. Cell Mol Life Sci 54,
46 Lionnet T, Czaplinski K, Darzacq X, Shav-Tal Y, 825–832.
Wells AL, Chao JA, Park HY, de Turris V, Lopez- 61 Calvin K & Li H (2008) RNA-splicing endonuclease
Jones M & Singer RH (2011) A transgenic mouse for in structure and function. Cell Mol Life Sci 65,
vivo detection of endogenous labeled mRNA. Nat 1176–1185.
Methods 8, 165–170. 62 Parker JS (2010) How to slice: snapshots of Argonaute
47 Park HY, Trcek T, Wells AL, Chao JA & Singer RH in action. Silence 1, 3.
(2012) An unbiased analysis method to quantify mRNA 63 Choudhury R, Tsai YS, Dominguez D, Wang Y &
localization reveals its correlation with cell motility. Wang Z (2012) Engineering RNA endonucleases with
Cell Rep 1, 179–184. customized sequence specificities. Nat Commun 3, 1147.
48 Querido E & Chartrand P (2008) Using fluorescent 64 Mackay JP, Font J & Segal DJ (2011) The prospects
proteins to study mRNA trafficking in living cells. for designer single-stranded RNA-binding proteins. Nat
Methods Cell Biol 85, 273–292. Struct Mol Biol 18, 256–261.
49 Yamada T, Yoshimura H, Inaguma A & Ozawa T 65 Nishikura K (2010) Functions and regulation of RNA
(2011) Visualization of nonengineered single mRNAs in editing by ADAR deaminases. Annu Rev Biochem 79,
living cells using genetically encoded fluorescent probes. 321–349.
Anal Chem 83, 5708–5714. 66 Singh RK & Cooper TA (2012) Pre-mRNA splicing in
50 Yoshimura H, Inaguma A, Yamada T & Ozawa T disease and therapeutics. Trends Mol Med 18, 472–482.
(2012) Fluorescent probes for imaging endogenous 67 Wang Y, Ma M, Xiao X & Wang Z (2012) Intronic
beta-actin mRNA in living cells using fluorescent splicing enhancers, cognate splicing factors and

3766 FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA.
Y. Wang et al. Designed PUF proteins

context-dependent regulation rules. Nat Struct Mol Biol stress: a matter of survival. Wiley Interdiscip Rev RNA
19, 1044–1052. 2, 336–347.
68 Ji Z & Tian B (2009) Reprogramming of 3′ 75 Adelman K & Lis JT (2012) Promoter-proximal
untranslated regions of mRNAs by alternative pausing of RNA polymerase II: emerging roles in
polyadenylation in generation of pluripotent stem cells metazoans. Nat Rev Genet 13, 720–731.
from different cell types. PLoS One 4, e8419. 76 Larochelle S, Amat R, Glover-Cutter K, Sanso M,
69 Sandberg R, Neilson JR, Sarma A, Sharp PA & Burge Zhang C, Allen JJ, Shokat KM, Bentley DL & Fisher
CB (2008) Proliferating cells express mRNAs with RP (2012) Cyclin-dependent kinase control of the
shortened 3′ untranslated regions and fewer microRNA initiation-to-elongation switch of RNA polymerase II.
target sites. Science 320, 1643–1647. Nat Struct Mol Biol 19, 1108–1115.
70 Thomsen S, Azzam G, Kaschula R, Williams LS & 77 Kotin RM, Siniscalco M, Samulski RJ, Zhu XD,
Alonso CR (2010) Developmental RNA processing of Hunter L, Laughlin CA, McLaughlin S, Muzyczka N,
3′ UTRs in Hox mRNAs as a context-dependent Rocchi M & Berns KI (1990) Site-specific integration
mechanism modulating visibility to microRNAs. by adeno-associated virus. Proc Natl Acad Sci USA 87,
Development 137, 2951–2960. 2211–2215.
71 Hall-Pogar T, Liang S, Hague LK & Lutz CS (2007) 78 Surosky RT, Urabe M, Godwin SG, McQuiston SA,
Specific trans-acting proteins interact with auxiliary Kurtzman GJ, Ozawa K & Natsoulis G (1997)
RNA polyadenylation elements in the COX-2 3′-UTR. Adeno-associated virus Rep proteins target DNA
RNA 13, 1103–1115. sequences to a unique locus in the human genome.
72 Newnham CM, Hall-Pogar T, Liang S, Wu J, Tian B, J Virol 71, 7951–7959.
Hu J & Lutz CS (2010) Alternative polyadenylation of 79 Li Y, Wang J, Satterle A, Wu Q & Liu F (2012) Gene
MeCP2: influence of cis-acting elements and trans- transfer to skeletal muscle by site-specific delivery of
acting factors. RNA Biol 7, 361–372. electroporation and ultrasound. Biochem Biophys Res
73 Peterson ML, Bingham GL & Cowan C (2006) Commun 424, 203–207.
Multiple features contribute to the use of the 80 Elzoghby AO, Samy WM & Elgindy NA (2012)
immunoglobulin M secretion-specific poly(A) signal but Protein-based nanocarriers as promising drug and gene
are not required for developmental regulation. Mol Cell delivery systems. J Control Release 161, 38–49.
Biol 26, 6762–6771. 81 Koren E & Torchilin VP (2012) Cell-penetrating
74 von Roretz C, Di Marco S, Mazroui R & Gallouzi IE peptides: breaking through to the other side. Trends
(2011) Turnover of AU-rich-containing mRNAs during Mol Med 18, 385–393.

FEBS Journal 280 (2013) 3755–3767 ª 2013. This article is a U.S. Government work and is in the public domain in the USA. 3767

You might also like