You are on page 1of 13

THE JOURNAL OF GENE MEDICINE RESEARCH ARTICLE

J Gene Med 2007; 9: 275–286.


Published online in Wiley InterScience (www.interscience.wiley.com) DOI: 10.1002/jgm.1014

Recombinant fusion proteins TAT-Mu, Mu and


Mu-Mu mediate efficient non-viral gene delivery

Rukkumani Rajagopalan Abstract


Jennifer Xavier
Nandini Rangaraj
Background The inherent ability of certain peptides or proteins of viral,
Nalam Madhusudhana Rao prokaryotic and eukaryotic origin to bind DNA was used to generate novel
Vijaya Gopal* peptide-based DNA delivery protocols. We have developed a recombinant
approach to make fusion proteins with motifs for DNA-binding ability,
Centre for Cellular and Molecular
Mu and membrane transduction domains, TAT, and tested them for
Biology, Uppal Road, Hyderabad
500007, India their DNA-binding, uptake and transfection efficiencies. In one of the
constructs, the recombinant plasmid was designed to encode the Mu moiety
*Correspondence to: Vijaya Gopal, of sequence MRRAHHRRRRASHRRMRGG in-frame with TAT of sequence
Centre for Cellular and Molecular YGRKKRRQRRR to generate TAT-Mu, while the other two constructs, Mu and
Biology, Uppal Road, Hyderabad Mu-Mu, harbor a single copy or two copies of the Mu moiety.
500007, India.
E-mail: vijaya@ccmb.res.in
Methods Recombinant his-tag fusion proteins TAT-Mu, Mu and Mu-Mu
were purified by overexpression of plasmid constructs using cobalt-based
affinity resins. The peptides were characterized for their size and interaction
with DNA, complexed with plasmid pCMVβ-gal, and shown to transfect
MCF-7, COS and CHOK-1 cells efficiently.

Results Recombinant fusion proteins TAT-Mu, Mu and Mu-Mu were cloned


and overexpressed in BL21(DE3)pLysS with greater than 95% purity. The
molecular weight of TAT-Mu was determined by matrix-assisted laser
desorption/ionization time-of-flight mass spectrometry (MALDI-TOFMS) to
be 11.34 kDa while those of Mu and Mu-Mu were 7.78 and 9.83 kDa,
respectively. Live uptake analysis of TAT-Mu, Mu and Mu-Mu as DP
(DNA+peptide) or DPL (DNA+peptide+lipid) complexes into MCF-7 cells,
followed by immunostaining and laser scanning confocal microscopy,
demonstrated that the complexes are internalized very efficiently and
localized in the nucleus. DNA : peptide complexes (DP) transfect MCF-7,
COS and CHOK-1 cells. The addition of cationic liposomes enhances the
uptake of the ternary complexes (DPL) further and also brings about 3–7-fold
enhancement in reporter gene expression compared to DP alone.

Conclusions Recombinant proteins that are heterologous fusions, having


DNA-binding domains and nuclear localization epitopes, generated in this
study have considerable potential to facilitate DNA delivery and enhance
transfection. The domains in these fusion proteins would be promising in the
development of non-viral gene delivery vectors particularly in cells that do
not divide. Copyright  2007 John Wiley & Sons, Ltd.
Received: 20 October 2006
Revised: 30 December 2006 Keywords gene delivery; non-viral gene delivery; cationic peptides; transfection;
Accepted: 15 January 2007 cationic liposomes; transfection; TAT-Mu; recombinant DNA; gene expression

Copyright  2007 John Wiley & Sons, Ltd.


276 R. Rajagopalan et al.

Introduction recognize and bind them. The use of such sequences in the
preparation of transfection complexes may therefore facil-
The natural process of DNA uptake by eukaryotic cells itate transport of the complexes efficiently to the nucleus
is inefficient owing to the chemical nature of the gene where gene expression must eventually occur [23], which
and the cell membrane. Invariably, gene delivery has to is the major rate-limiting step [10,24]. Transfection-
be facilitated by carriers both viral and non-viral with enhancing peptides, synthesized by solid-phase synthesis,
examples of cationic lipids [1,2] and polymers [3,4] in have reportedly shown reporter gene expression in various
the non-viral category. These are known to interact with cell lines.
DNA resulting in the formation of condensed, transfection- Our strategy in this study aims at designing and pro-
competent complexes. Gene delivery by non-viral methods ducing small proteins encoded by plasmid constructs by
is less efficient in cells where the mitotic rates are low. conventional recombinant DNA strategies and bacterial
Cells that divide slowly or differentiated cells are often fermentation rather than the synthetic route. A basic
resistant to current DNA delivery methods. Understand- domain of TAT was earlier shown to be the minimal
ing the mechanism of nucleic acid uptake by cells coupled sequence responsible for the cellular and nuclear uptake
with the development of methods to make this an efficient which is contributed by potential nuclear localization
process forms the major activity of research in the area of sequences (NLSs) in the 11-amino acid epitope YGRKKR-
gene delivery. Although gene transfer methods based on RQRRR [25]. The TAT moiety has also been shown to
viruses are successful, challenges such as immunogenicity deliver functional fusion proteins in vivo [26]. By com-
[5], toxicity [6] and their ability to integrate into the host bining the Mu motif with TAT, the inherent, desired
genome [7] have to be overcome. Cationic amphiphiles properties, such as DNA condensation of Mu and nuclear
have been widely employed for in vitro transfections as localization conferred by TAT, facilitation of transport
these offer many advantages over that of viral systems in across plasma and nuclear membrane, could be integrated
terms of the ease of production, ability to carry larger DNA into one molecule. Here, we report the generation of the
transgenes, stability and safety in clinical trials [2,8]. In plasmid constructs that overproduce TAT-Mu, Mu, and
the past decade, the optimization of conditions for trans- Mu-Mu as heterologous, his-tag fusion proteins. We eluci-
fection has taken a rather long route as a large number date their DNA-binding, uptake and transfection potential
of these non-viral delivery agents and their derivatives into cells. Recombinant fusion proteins TAT-Mu, Mu and
are initially tested empirically for their efficiency. A more Mu-Mu have the potential to bind plasmid DNA, increase
rational approach is to address the issues concerning vari- internalization of transfection complexes into adherent
ous barriers and pathways leading eventually to the entry cells in culture, and thereby enhance transfection.
of DNA or transfection complexes of DNA into the nucleus,
which is the ultimate barrier in the transfection process.
This is elegantly discussed in recent reviews pertaining to Materials and methods
intracellular trafficking of nucleic acids, non-viral vectors
and polyplexes [9,10] that forms a major part of non-viral Chemicals
gene delivery strategies.
1,2-Dioleoyl-3-trimethylammonium propane (DOTAP),
Cationic peptides that condense DNA have a tremen-
3 beta-(N(N  ,N-dimethylaminoethane)carbamoyl) choles-
dous potential to facilitate the passage of DNA through
terol (DC-Chol) and dioleoylphosphatidylethanolamine
various mechanisms [11–17] thereby circumventing
(DOPE) were purchased from Avanti Polar Lipids Inc.
cell membrane barriers. The incorporation of pep-
N-(Lissamine rhodamine B sulfonyl)-1,2-dihexadecanoyl-
tides in transfection protocols with DNA-binding poten-
sn-glycero-3-phosphoethanolamine (Rh-DHPE) was from
tial/nuclear localization motifs has been shown to
Molecular Probes. Plasmid pTAT was a kind gift from
improve the transfection efficiency [14,15,18–20] and
Steve Dowdy’s laboratory (UCSD). Plasmid pHind2 was
may offer solutions to deliver DNA more effectively.
a kind gift from Akusjarvi of Uppsala University Sweden.
Several adenoviral proteins such as the Mu, with pre-
Plasmid DNA pCMV.SPORT-β-gal was from Invitrogen.
dominantly basic amino acid residues, have the potential
BD TALON was from BD Biosciences. The primary 6xhis
to bind DNA and also bring about enhancement in cationic
monoclonal antibody was from BD Biosciences. FITC
lipid-mediated transfection [13,19]. Mu (µ) is based on
secondary antibodies were from Bangalore Genei. Flu-
the adenoviral core peptide of 19 amino acids with a
orescein DNA-labeling kits for labeling plasmid DNA by
sequence ‘MRRAHHRRRRASHRRMRGG’, twelve of which
nick translation and random primer labeling were from
are basic, conferring positive charge on the protein. This
Jonaki, India. Ethidium bromide (EtBr) was from Sigma
peptide is found associated with the adenoviral core com-
Aldrich. All other chemicals used were of the highest
plex [21,22]. Work in Miller’s laboratory has shed light
purity available.
on using systems that are based on the DNA-condensing
ability of synthetic Mu peptide and has also shown its
ability to enhance cationic lipid-mediated transfections PCR amplification, sub-cloning
in vitro [13].
The movement of macromolecules within cells is often The Mu moiety from the plasmid pHind2 was amplified
aided by nuclear localization sequences and factors that using forward and reverse primers with restriction

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
Recombinant Fusion Proteins Mediate Efficient Gene Delivery 277

sites KpnI and EcoRI incorporated in them. This was DNA-EtBr complex and this displacement of EtBr by the
subsequently cloned in-frame into the pTAT construct fusion proteins is reflected as a drop in the fluorescence
similarly restricted to generate pTAT-Mu that is inducible signal. EtBr from a stock solution of 1 mg/ml was added to
in the presence of 1 mM IPTG when expressed in DNA in a cuvette and the fluorescence was measured using
an overexpression host BL21(DE3)pLysS. In the Mu a fluorescence spectrophotometer (Hitachi-F4000). The
construct, a BamH I digestion of pTAT-Mu followed by excitation wavelength, λex , was 516 nm and the emission
recircularization has deleted the TAT moiety. Inserting a wavelength was kept at 598 nm (slit width 5 × 5 nm).
second copy of Mu amplified by polymerase chain reaction Briefly, 2.3 µg of pCMV-β-gal plasmid DNA was added to
(PCR) into the Mu construct generated the Mu-Mu 500 µl of sample in 20 mM Tris-HCl buffer (pH 7.4) in
plasmid. These constructs, pTAT-Mu, pMu and pMu-Mu, a fluorescence cuvette. EtBr (0.23 mg) was added to the
were verified for the integrity of the DNA insert sequence DNA solution and the baseline fluorescence was deter-
prior to transformation in E. coli. The plasmids were then mined. The fluorescence intensity obtained upon each
transformed into competent E. coli BL21(DE3)pLysS cells, addition was normalized relative to the fluorescence sig-
grown and induced to express the desired protein. nal of the DNA-EtBr complex in the absence of the fusion
proteins, which was taken as 100%. The binding of DNA
by the fusion peptides TAT-Mu, Mu and Mu-Mu, respec-
Overexpression and purification of the tively, was recorded after each addition at time intervals
recombinant proteins of 5 min. The binding efficiency correlates with a drop in
the fluorescence intensity at various time points.
Recombinant TAT-Mu, Mu and Mu-Mu proteins were
purified from bacterial pellets by urea denaturation fol-
lowed by the use of cobalt affinity matrix (BD TALON; Preparation of complexes for
BD Biosciences). This matrix eliminates the binding of transfection
endogenous contaminating E. coli histidine-rich protein
that co-purifies with our protein in a nickel-affinity matrix, Cationic liposomes DOTAP and DC-Chol were prepared
thereby enriching the protein of our interest during elu- by drying the appropriate amount from their chloroform
tion. PD-10 desalting columns subsequently removed urea stock solutions. Plasmid DNA pCMVβ-gal was purified by
employed as a denaturant in the purification protocols. using the Qiagen kit (endotoxin free) using the manu-
The final yield was typically 2 mg from a 1000 ml culture facturer’s protocol. Complexes were prepared by taking
where the protein was greater than 95% homogeneous, 0.9 µg of plasmid DNA and varying amounts of the fusion
as ascertained by sodium dodecyl sulfate/polyacrylamide protein to obtain the desired charge ratio, in Hepes
gel electrophoresis (SDS-PAGE). Western blotting of these buffered saline (pH 7.4), and incubated for 25 min at
purified proteins was carried out using monoclonal anti- RT. Transfection experiments were initially performed
his antibodies against the 6xhis-tag fusion proteins fol- with the TAT-Mu, Mu and Mu-Mu peptides and DNA
lowed by binding to the secondary antibody. complexes. In liposome-mediated transfections, cationic
liposomes were added to the pre-incubated DNA : peptide
DNA binding and electrophoretic complexes and further incubated for 25 min at RT. This
ternary complex was then diluted with serum-free Dul-
mobility shift assays
becco’s modified Eagle’s medium (DMEM) to a final
Charge neutralization and DNA condensation of the volume of 300 µl before splitting them into triplicates
fusion peptides were observed by electrophoresis of of 100 µl volume on a 96-well plate format containing
the complexes on agarose gels. Plasmid DNA pEGFPN3 20 000 cells per well. The cells were incubated for at least
(0.2–0.4 µg) was complexed with increasing amounts 3 h following which complete medium with serum was
of the TAT-Mu/Mu/Mu-Mu peptide, to obtain the corre- added and the reporter gene expression was assayed for
sponding charge ratios, in Hepes buffered saline (150 mM β-galactosidase 24–48 h post-transfection.
NaCl, 5 mM KCl, 0.75 mM Na2 HPO4, 20 mM Hepes pH
7.4) and incubated for 25 min at room temperature (RT).
Live uptake and immunolocalization of
The complexes were subsequently analyzed on a 1%
agarose gel [Tris-acetate buffer (TAE) system, pH 8.2]
peptide : DNA complexes by confocal
and stained with EtBr post-electrophoresis. microscopy

MCF-7 cells were grown on either a chambered cover


EtBr exclusion assay glass (Lab-Tek, Nalgene, Nunc International Corp.) or
on cover slips placed on six-well plates. Cells plated to
The binding of DNA with TAT-Mu, Mu and Mu-Mu was ∼70% confluence were washed with serum-free DMEM
studied by monitoring the fluorescence with the fluores- (pH 7.4) and then incubated with the complexes at 1 : 4
cent probe EtBr. The intercalation of EtBr into DNA brings DNA : peptide charge ratio, at 37 ◦ C and 5% CO2 . In the
about an increase in the fluorescence quantum yield. Upon case of fluorescent cationic lipid, DOTAP was labeled
binding and condensing DNA, EtBr is expelled from the with 5 mol% Rh-DHPE. For the live uptake experiments,

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
278 R. Rajagopalan et al.

plasmid DNA was labeled (at 1 : 1 ratio of labeled vs. with an apparent molecular weight that is slightly larger
unlabeled DNA) by either the random primer labeling than that deduced, based on the DNA sequence of the
kit or by the nick translation kit using fluorescein-d UTP plasmid inserts. The recombinant TAT fusion proteins
from Jonaki, combined with the peptide and incubated migrated slower than expected with an apparent increase
at 37 ◦ C. Subsequently, these were examined by confocal of 7–10 kDa on SDS gels. This apparent difference could
microscopy to monitor the movement of the complexes be attributed to both the TAT leader sequences as well
into cells. The image acquisition conditions for all the as the highly cationic nature of the protein. That the
experiments were identical. TAT leader sequences contribute to ∼5 kDa increase in
The immunolocalization experiment was similar to the molecular weight has been reported for other TAT fusion
live uptake experiment except that, after the incuba- proteins [27]. The presence of the his-tag of the fusion
tion process, cells were exposed to anti-his antibodies proteins was confirmed by immunoblotting using anti-his
directed against the 6xhis-tag fusion proteins after uptake. antibodies that confirmed the authenticity of the proteins
After incubation for 1 h, the cells were fixed with 2% (inset of Figure 1B, lanes 1–3). The corresponding molec-
paraformaldehyde, permeabilized with 0.25% Triton X- ular masses for TAT-Mu, Mu and Mu-Mu, as derived by
100 followed by immunostaining with anti-his antibodies, matrix-assisted laser desorption/ionization time-of-flight
at a dilution of 1 : 200, and incubated overnight at RT. mass spectrometry (MALDI-TOFMS), are 11.34, 7.78 and
This was followed by the addition of propidium iodide 9.83 kDa, respectively.
(PI) with the secondary antibody FITC-conjugated goat
anti-mouse IgG (anti-mouse used at a dilution o f 1 : 1000
and then analyzed with a confocal microscope (LSM 510 DNA-binding analysis
META, Carl Zeiss). Optical sections of cells were taken
using a 63× objective at 0.5 µm intervals. A 518 nm Gel retardation assay
laser line was used to excite the FITC fluorophore in the We next investigated the ability of recombinant proteins
secondary antibody and a 543 nm laser to visualize the TAT-Mu, Mu and Mu-Mu to bind plasmid DNA by an
rhodamine fluorophore. Images were collected simulta- agarose gel retardation assay. Plasmid pEGFPN3 , harbor-
neously for PI staining of the nuclei. The images were ing the gene encoding the green fluorescent protein (GFP),
analyzed by taking equal numbers of optical sections in was used for the binding assay. The number of arginines
the central region and then merged. and lysines in the fusion protein were taken into account
in order to calculate the total positive charges on the
molecule. Complexes of DNA and protein were prepared
Results at various charge ratios and incubated with the plasmid
at 37 ◦ C for 15 min and electrophoresed. In the absence
of protein, which is the control, both relaxed and super-
Construction and purification of
coiled DNA migrated normally. At charge ratios of 1 : 8
multifunctional DNA carrier proteins and 1 : 16 (DNA : protein), the ability to bind DNA was
TAT-Mu, Mu and Mu-Mu the highest for Mu-Mu while both TAT-Mu and Mu had
similar binding patterns, as seen from the shift in the
The DNA-binding domain of Mu was derived as described mobility of plasmid DNA. Both relaxed and supercoiled
in the ‘Materials and methods’ section by restriction forms of DNA were retarded by their interaction with
digestion, PCR amplification and assembly in-frame in the fusion proteins (Figure 1C, panel a – TAT-Mu; panel
the expression construct pTAT to obtain pTAT-Mu. The b – Mu; panel c – Mu-Mu). In the case of Mu-Mu at 1 : 8
pMu construct was obtained by BamH I excision of the charge ratio, the binding to EtBr is occluded as seen from
TAT moiety of pTAT-Mu plasmid. In the case of Mu- the decreased DNA band intensity. At 1 : 16 charge ratio,
Mu, an additional copy of the Mu moiety was cloned the DNA appears to be inaccessible to EtBr binding. The
in-frame into the pMu construct. All the three plasmid ability of the peptides to protect DNA in the complex from
constructs encoding the fusion protein are under the con- nucleases was investigated by incubating the complexes
trol of the T7 promoter with an N-terminal histidine tag. with DNase I. Protection from nucleases was complete at
Figure 1A shows the schematic of the fusion constructs charge ratios of 1 : 8 (data not shown).
with the placement of 6xhis-tag at the N-terminus. The
presence of the his-tag in-frame with TAT-Mu, Mu and
Mu-Mu facilitates the purification of the recombinant pro- Displacement of EtBr from DNA by TAT-Mu, Mu and
teins using cobalt affinity matrix. The fusion proteins Mu-Mu: DNA-binding assay
were expressed in E. coli BL21(DE3)pLysS. Total bacterial Titrating DNA-EtBr complexes with the fusion proteins
lysates were obtained with 8 M urea and the recombi- further elucidated the differential DNA-binding charac-
nant protein purified by elution with 250 mM imidazole. teristics. In this experiment, the addition of the fusion
The fractions were analyzed on 15% SDS gels, pooled proteins to the DNA-EtBr complexes resulted in a rapid
and then the denaturant and imidazole were removed decrease in the fluorescence intensity (Figure 1D). The
with PD-10 columns. Figure 1B shows an SDS-PAGE anal- binding efficiency of the fusion proteins correlates with a
ysis of the purified proteins appearing as single bands drop in the fluorescence intensity and this loss could be

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
Recombinant Fusion Proteins Mediate Efficient Gene Delivery 279

interpreted as their ability to bind DNA that results in the the DNA is condensed appears to be different in the case
displacement of the intercalated EtBr. Although the three of Mu-Mu, which is the most efficient, while TAT-Mu
fusion proteins were able to bind DNA, as seen from the and Mu displayed similar DNA-binding affinity. When we
gel binding assays depicted in Figure 1C, the fluorescence monitored the binding properties of TAT, we observed
titrations clearly indicate that the efficiency with which that TAT does not have any effect on EtBr exclusion even
at 1 : 8 charge ratio indicating its lack of binding to DNA
(not shown).

TAT-Mu, Mu and Mu-Mu mediate


transfer of DNA into cells

Live uptake
Having ascertained the DNA-binding characteristics of the
fusion proteins, the internalization of DNA : peptide (DP)
or DNA : peptide : lipid (DPL) complexes into the cells
was investigated by monitoring the live uptake of the
complexes by confocal laser scanning microscopy. After
the application of complexes of DP or DPL, cells were
incubated in the absence of serum for 1 h and replaced
with Hepes/Hanks buffer containing 10% serum. In the
case of the DP complexes, nick translated plasmid DNA
was combined with the fusion proteins and the uptake
into MCF-7 cells was visualized live. The localization of
DP complexes appeared as punctate particles in both the
cytoplasm and the nucleus. It is evident that the DP
complexes, D + TM, D + Mu and D + Mu-Mu, prepared
with TAT-Mu, Mu and Mu-Mu, respectively, facilitate the

Figure 1. Construction and gel analysis of DNA-binding carrier


proteins. (A) Schematic representation of chimeric fusion
constructs encoding (a) TAT-Mu, (b) Mu, and (c) Mu-Mu. The
motifs harboring the TAT sequence YGRKKRRQRRR and/or Mu
sequence MRRAHHRRRRASHRRNRGG are cloned in-frame in a
pTAT expression vector and expressed as N-terminal his-tag
fusion proteins in E. coli. The plasmids are designated as
pTAT-Mu, pMu and pMu-Mu. The Mu construct is created by
a deletion of the TAT moiety of the plasmid construct pTAT-Mu
as described in the ‘Materials and methods’ section. (B) Sodium
dodecyl sulfate/polyacrylamide gel electrophoretic (SDS-PAGE)
analysis of the purified fusion proteins. Lane 1, 2, 3 correspond
to TAT-Mu (2 µg), Mu (∼2 µg) and Mu-Mu (1 µg) after desalting
from PD-10 columns. The molecular weights of the expressed
proteins TAT-Mu, Mu and Mu-Mu are 11.34, 7.78 and 9.23 kDa,
respectively. Inset shows the immunoblot analysis of purified
proteins TAT-Mu, Mu and Mu-Mu after electrophoresis and
Western transfer. The 6xhis-tagged proteins were detected with
monoclonal anti-his antibodies followed by secondary antibody
goat anti-mouse IgG AP conjugate. Detection was by NBT/BCIP
treatment. M, molecular weight standards in kDa. (C) Gel
retardation assay of the fusion proteins TAT-Mu, Mu and Mu-Mu.
In each panel, i.e. (a)–(c), plasmid pEGFPN3 (0.4 µg in (a) and
0.2 µg in (b) and (c)) was incubated in 1× Hepes buffered
saline (pH 7.4) with increasing amounts of the purified protein
corresponding to the charge ratio indicated above each lane.
Samples were electrophoresed on 1% agarose gel in TAE buffer
and visualized by staining with ethidium bromide (EtBr). Control
in each panel pertains to plasmid DNA alone. (D) DNA binding
of TAT-Mu, Mu and Mu-Mu to DNA: Titration curves depicting
the release of EtBr from pCMVβ-gal plasmid DNA upon binding
TAT-Mu (solid squares), Mu (solid triangles) and Mu-Mu (open
circles) fusion proteins in a buffer containing 10 mM TrisHCl
(pH 7.4). 8 µg of protein corresponds to approximately 1 : 2.5
charge ratio (DNA : protein). Details as in the ‘Materials and
methods’ section

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
280 R. Rajagopalan et al.

Figure 2. Live uptake of transfection-competent complexes into MCF-7 cells. (A) Confocal microscope analysis of uptake of
FITC-labeled DNA + fusion protein complexes into MCF-7 cells. Panels from left to right: Control: DNA alone; DNA : TAT-Mu;
DNA : Mu; DNA : Mu-Mu all at 1 : 4 charge ratio DNA : protein. 0.9 µg of DNA was used to make the transfection complexes with
the corresponding protein and applied to cells in the absence of serum. Uptake of the complexes is for a duration of 3 h before
replacement with complete medium containing 10% serum. Thirty minutes before the confocal analyses, the cover slips were placed
in Hepes/Hanks buffer. (B) Confocal microscope analysis of uptake of DNA + fusion protein + lipid complexes into MCF-7 cells.
Complexes were prepared using rhodamine-labeled DOTAP liposomes (5 mol% Rh-DHPE) and the uptake experiment was carried out
as described in the ‘Materials and methods’ section. Panels on the left show the images in the fluorescence channel and those on the
right show the images merged with the transmission channel. Eight optical sections in the middle region, each of 0.5 µm thickness,
were combined to generate the images. D + L = DNA:liposomes at 1 : 1 charge ratio, D + TM + L = DNA + TAT-Mu + liposomes
(1 : 4 : 1), D + Mu + L = DNA + Mu + liposomes (1 : 4 : 1 charge ratio) and D + Mu-Mu + L = DNA + Mu-Mu + liposomes (1 : 4 : 1)

uptake of DNA into the cytoplasm and the nucleus when intensity (Figure 2B). In a situation where both labeled
compared to the control DNA (extreme panel on the DNA and rhodamine-labeled lipid were used to prepare
left) that is labeled DNA alone (Figure 2A). Both nick the DPL complexes, with the respective proteins, the
translation and random primer labeling methods to label presence of a dual label, originating from the FITC-labeled
plasmid DNA yielded identical end results. plasmid DNA as well as the rhodamine-labeled lipid,
In the case of live uptake of DNA : peptide : lipid within cells indicated that the DNA and lipid co-localized
complexes (DPL), cationic lipid DOTAP was labeled with (not shown).
5 mol% Rh-DHPE. The punctate red fluorescence (seen
in all panels) was observed upon uptake and entry Immunocytochemistry of TAT-Mu, Mu and Mu-Mu fusion
of the particles into the cytoplasm. When compared proteins
to DL (DNA + lipid) complexes, the uptake of DPL In yet another set of experiments, we also monitored
complexes prepared with the three fusion proteins the intracellular localization of the complexes after an
was enhanced as seen from the increased fluorescence incubation period of 1 h followed by immunolocalization

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
Recombinant Fusion Proteins Mediate Efficient Gene Delivery 281

Figure 3. Immunolocalization and confocal microscopy of fusion proteins. Fusion proteins : plasmid pCMVβ-gal DNA complexes
(DP) prepared with TAT-Mu (a), Mu (b), and Mu-Mu (c) at 1 : 4 charge ratio and incubated for 1 h with cells at 37 ◦ C. After
incubation, cells were fixed followed by immunostaining with anti-his antibodies followed by propidium iodide (PI) addition with
the FITC-conjugated secondary antibody goat anti-mouse IgG. The localization of the his-tagged proteins in the cytoplasm and the
nucleus is visualized as green fluorescence while PI staining of the nucleus is in red. Equal numbers of optical sections in the
middle region, each of 0.5 µm thickness, were combined to generate panels in (a)–(f). Top panels correspond to DP complexes
alone incubated with MCF-7 cells for 1 h while the bottom panels (d–f) correspond to TAT-Mu, Mu and Mu-Mu, respectively, to
which DPL complexes prepared with DC-Chol : DOPE at 1 : 4:1 charge ratios were applied to cells and incubated for 1 h. Details as
described in the ‘Materials and methods’ section

with FITC-labeled secondary antibodies (Figure 3). We in the ‘Materials and methods’ section. We compared
observed that the entry of complexes into the cytoplasm the transfection efficiency of the transfection complex
and nucleus occurs rapidly within 1 h, as seen from the at the optimized charge ratio of 1 : 8. We chose the
green fluorescence in the cytoplasm and the nucleus optimized charge ratio of 1 : 1 DNA : lipid as in our
in the case of DP complexes prepared with TAT-Mu, hands DDAB : DOPE, Lipofectin and also DC.Chol : DOPE
Mu or Mu-Mu (Figures 3a–3c). The nuclear staining is gave the best results. Where complexes (DPL) were
very intense in all the panels where cells were treated prepared with the fusion proteins and the cationic lipid,
with DPL complexes prepared with DC-Chol : DOPE we obtained enhanced transfection at 1 : 8:1 charge
(Figures 3d–3f). Panels a and b in the Supplemental ratio of D : P : L. Figures 4a–4c show a representative
Section S1 correspond to without-primary antibody set of the results obtained in MCF-7 cells. Complexes
control and cell control, respectively (see Supplementary of pCMVβ-gal : fusion protein DP, i.e. D + TM, D + Mu or
Material). D + Mu-Mu, at 1 : 8 charge ratio, were very effective at
transfecting cells when compared to plasmid pCMVβ-gal
alone. Lipopolyplex mixtures DPL prepared with TAT-
TAT-Mu, Mu and Mu-Mu transfection in MCF-7 cells Mu (D + TM + L) resulted in ∼7.5-fold enhancement
Having ensured the DNA-binding and uptake properties in transfection when compared to D + TM and 3.5-
of DP and DPL complexes prepared with the recombinant fold increase was observed when compared to D + L
fusion proteins TAT-Mu, Mu and Mu-Mu, we went on (Figure 4a). Similar fold increases were obtained for TAT-
to determine the relative transfection efficiencies of Mu with the cationic lipid Lipofectin or DDAB : DOPE (not
fusion proteins. Transient transfections during the initial shown).
characterization of the fusion proteins TAT-Mu, Mu and In the case of the fusion protein Mu, ∼4-fold
Mu-Mu were tested at various charge ratios varying from increase in the transfection efficiency with D + Mu + L
1 : 2 to 1 : 16. We have consistently obtained maximum (L = DC-Chol : DOPE) was obtained when compared with
transfection when cells were transfected with complexes D + Mu complexes. The fold increase in transfection
at 1 : 8 charge ratio of DNA : fusion protein with TAT-Mu, efficiency obtained with D + Mu + L prepared with DC-
Mu or Mu-Mu (not shown). Chol : DOPE is ∼3-fold when compared with that of
After the initial characterization, transient transfections D + L (Figure 4b). We also obtained good transfection
were carried out in MCF-7 cells with plasmid pCMVβ- efficiencies with Mu-Mu fusion protein where D +
gal : lipid DL or DPL complexes prepared with TAT- Mu-Mu + L complexes resulted in ∼7-fold enhancement
Mu, Mu or Mu-Mu fusion proteins and reporter gene in transfection when compared to D + Mu-Mu alone
expression was assayed 48 h post-transfection, as detailed and ∼3-fold when compared with D + L. Complexes

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
282 R. Rajagopalan et al.

or DC.Chol : DOPE under the conditions tested (not


shown).
A charge ratio of 1 : 1 of DNA to lipid was generally
found to be optimal for several cationic lipids developed
in our laboratory and with those reported in the literature.
Unlike cationic lipids, higher charge ratios greater than
1 : 8, resulting from peptides in a polyplex, were non-toxic.
The effect of charge ratio in a lipoplex in transfection is
not similar. In the case of DPL complexes, a charge ratio
higher than 1 : 8:1 or lower than 1 : 4:1 always resulted in
less efficient transfection (not shown).
We also examined the transfection efficiency of a
control peptide such as poly-l-lysine in the presence and
absence of Lipofectin. Poly-l-lysine 3 kDa and 30 kDa
were both tested in MCF-7 cells and compared to a lipid-
DNA control under the same conditions. The transfection
efficiency with poly-l-lysine in the absence of lipid was
similar to the fusion peptides. The transfection efficiency
with the three fusion proteins was ∼5–8 times higher
when compared to poly-l-lysine in the presence of the
lipid (not shown).

Discussion
Non-viral vectors are comparatively much safer although
not as efficient as viral vectors [8]. It is vital to
develop non-viral vector systems and make them clinically
efficient. The use of peptides to overcome the cellular
barriers of DNA entry into the cell and then into the
nucleus is gaining momentum. When testing out gene
delivery systems, the cell membrane is the first major
barrier. An approach to overcome this problem is to
incorporate motifs with peptides that are capable of
penetrating the cell membrane. Subsequently, the delivery
of DNA from the cytoplasm into the nucleus, that is also an
inefficient process and considered to be the major barrier
in non-dividing cells, is challenging. A strategy to improve
nuclear uptake of DNA is to exploit the cellular nuclear
import machinery where peptides containing an NLS can
be conjugated and thereby facilitate the translocation
of macromolecules across the nuclear envelope inside
Figure 4. Transient transfection of MCF-7 cells with (a) TAT-Mu cells. Few synthetic peptides with DNA-binding ability or
(TM), (b) Mu, and (c) Mu-Mu. Complexes prepared with the nuclear import capabilities have been shown to enhance
fusion proteins and the lipids are at a charge ratio of 1 : 8:1 and transfection efficiency. In this report we have presented a
prepared as indicated in the ‘Materials and methods’ section.
strategy to make combinations of various peptide motifs
Protein estimation of the cell lysates was done to express
β-galactosidase activity in mU after normalizing the protein using recombinant genetic approaches and to produce
values. D-plasmid DNA alone (control); L-lipid DC-Chol : DOPE. them by heterologous expression. The approach allows
The lipids used were DC-Chol : DOPE in all panels at 1 : 1 the testing of various combinations of the peptides that
charge ratio. The reporter gene assay was carried out 48 h are efficient DNA carriers. Based on this, novel peptide-
post-transfection
based gene carriers could be designed for gene therapy
applications.
of the fusion proteins and plasmid DNA harboring the Our efforts to identify potential cationic proteins for
β-galactosidase or GFP gene also transfect COS, HeLa transfection led to the construction of plasmids that
and CHOK1 cells efficiently both in the absence and encode the fusion proteins TAT-Mu, Mu and Mu-Mu that
presence of cationic liposomes (data not shown). Purified are cationic in nature. Mu is characterized by the 19-amino
TAT as such did not result in any enhancement in acid epitope, rich in arginines, and potential to condense
transfection either in the absence or presence of Lipofectin the viral DNA [21]. While Mu helps condense DNA,

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
Recombinant Fusion Proteins Mediate Efficient Gene Delivery 283

the short, basic peptide sequence from the HIV-1 TAT of peptides rich in basic amino acids, in bacterial systems,
protein known as TAT or protein transduction domain usually results in low yields and in our hands required
(PTD) (residue 47–57) confers the property of cellular the standardization of protocols to further improve
uptake across membranes and nuclear localization. As the yields. In our experience, the fusion proteins are
evident from our results, the single and double copies of present as insoluble fractions and require denaturation
the Mu moiety in the recombinant Mu and Mu-Mu are followed by renaturation to obtain pure preparations.
as efficient as TAT-Mu in bringing about reporter gene To aid in purification of the peptides, DNA sequences
expression although the DNA-binding pattern of Mu-Mu corresponding to the peptides were cloned downstream
is different from that of TAT-Mu and Mu. Interestingly of the his-tag present at the N-terminus. The presence of
the transfection efficiency of these DP complexes was the his-tag at the N-terminus enabled us to confirm the
further enhanced when the transfection complexes were purity of the peptide preparation by Western blotting.
formulated with cationic liposomes, i.e. DPL. These fusion In addition, the presence of histidine residues in a
proteins have enhanced the transfection of efficient transfection complex is known to bring about osmotic lysis
and well-studied cationic lipid formulations of which of endosomes upon protonation due to acidification of
only results obtained with DC-Chol : DOPE have been endosomes [28] conferring a broad utility of these fusions
presented here. in gene therapy applications. All the three fusion proteins,
Although several factors such as the primary sequence, TAT-Mu, Mu and Mu-Mu, have DNA-binding properties
net charge and sequence context may all be important, and also the ability to protect the DNA from nucleases. The
a combination of these could possibly influence gene charge ratio required for retardation of the plasmid DNA
transfer in a positive manner. The presence of the TAT and protection from nuclease occurs at 1 : 8 DNA : protein,
motif in TAT-Mu may thereby synergistically facilitate indicating complete occlusion of the plasmid by peptides.
the import of plasmid DNA into the nucleus, as it Within 1 h of addition of DP, extensive internalization of
has better DNA-binding ability than TAT alone. These peptides along with the plasmid DNA was observed. The
properties finally relate to the enhanced transfection rapid entry of DP and particulate fluorescence suggests
observed for TAT-Mu both in the absence and presence that the endocytic pathway mediates the uptake of the
of cationic lipids. Hyndman et al. have observed the complexes. The cellular entry of DP or DPL complexes
unexpected negative effect of also tagging TAT with was confirmed by using fluorescent probes where both
poly-l-lysine [19] which indicates that the primary DNA and the lipid were shown to co-localize. Based on
sequence of TAT may play a role in determining peptide- intensity of fluorescence as shown by immunostaining and
mediated enhancements in the transfection efficiency in a fluorescence-based DNA binding assay, it is apparent
the presence of liposomes. From the results with poly- that Mu-Mu shows maximum internalization even in the
l-lysine, we reasoned that a stretch of positive charges absence of lipid. A significant amount of fluorescence was
in the cationic polymer may not be as efficient as also seen inside the nucleus suggesting that in 1 h DP or
peptides described in this study. The presence of multiple DPL complexes have entered the nucleus. In comparison
arginines in the double copy of the Mu-Mu construct to this, Mu apparently shows more internal fluorescence.
may also explain the stronger binding to DNA as seen by When the internal fluorescence was monitored using the
immunolocalization. Nuclear import of nucleic acids may his-tag antibodies, the results showed much stronger
be inefficient during cationic lipid-mediated transfection. fluorescence inside the nucleus of MCF-7 cells. The
A number of attempts to improve the transfection intensity was more pronounced with DPL than DP. Though
efficiency by attempting to enhance nuclear uptake of significant particulate fluorescence is seen with DP or
nucleic acids by using polycationic polymers bearing DPL in fixed samples strong fluorescence seen inside the
peptide sequences with nuclear localizing capabilities nucleus may be partially attributed to the process of
have met with success. The presence of NLSs does fixation per se. During fixation, the cationic peptides may
not promote transfection without adequate nucleic acid access the nuclear DNA and bind avidly thereby resulting
charge neutralization and condensation as well. For in strong fluorescence. However, the results obtained
example, synthetic TAT alone complexed with DNA was with live imaging of DP uptake using FITC-labeled DNA
unable to transfect cells but facilitated transfection in and co-localization experiments clearly indicated that
the presence of cationic liposomes [19]. The domains in these peptides have facilitated rapid entry of the DP
TAT-Mu provide the attributes important in contributing or DPL into the MCF-7 cells. Efficient internalization of
the individual functions and may lay the foundation for plasmid DNA was supported by our data that the fusion
designing next-generation constructs where issues such proteins are able to bring about reporter gene expression
as the binding strength and nuclear localization potential in MCF-7 cells (∼100-fold) when compared to plasmid
need to be considered. In the early studies, with cationic pCMVβ-gal DNA alone. Between the three fusions, the
peptides in transfection, the peptides were generated by efficiency of transfection was enhanced in the presence
solid-phase synthesis. Such labor-intensive methods of of DC-Chol : DOPE contributing to 3–7-fold differences
peptide synthesis limit testing of larger peptides and also between DP and DPL for TAT-Mu, Mu and Mu-Mu
peptide combinations. To circumvent these issues, few fusion proteins. It has been demonstrated that cationic
laboratories have attempted to synthesize peptides using polymers such as poly-l-lysine and protamine enhance
recombinant DNA approaches. Heterologous production lipid-mediated transfections in several cell lines in vitro

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
284 R. Rajagopalan et al.

[29]. Similarly, several DNA-condensing natural peptide example, subsequent modification was very essential
sequences including Mu [13], HMG [30], and histones to increase its utility in gene delivery. A stretch of
[31], etc., have been demonstrated to enhance reporter positive charges alone may not be sufficient for enhancing
gene expression when included in the transfection transfection. Also, the presence of nuclear localization
protocols. A recent study from Wels’ laboratory has shown does not promote transfection without adequate nucleic
that recombinant derivatives of human HMGB2 facilitate acid charge neutralization and condensation as well.
non-viral gene delivery [15]. Yet another study used a In all the reported instances, the inclusion of peptides or
similar approach by generating a recombinant polymer- cationic polymers has resulted in more uniform complexes
fusion protein (KH)-FGF2 that had the ability to condense of 100–200 nm size compared to much larger particles
DNA and mediate transgene expression in mammalian that result from complexes prepared with cationic lipids.
cells [32]. We have also observed that the size of DPL is ∼150 nm
Synthetic Mu was extensively investigated for its ability compared to a few hundreds of nanometers in the
to enhance transfection efficiency in post-mitotic neuronal case of DL prepared with DC-Chol : DOPE and detailed
cells and also in vivo lung transfection. These studies investigations are underway. Though it is expected
have led to LMD formulations containing lipids and Mu that the cellular uptake of the DPL is mediated by
peptide which are efficient transfection formulations at endocytosis, it is not very clear how various components
very low amounts of DNA and in the presence of 100% of DPL interact with various organelles inside the cell.
serum [13]. Examination of peptide sequences of the Preliminary transfection data obtained by the addition of
fusion proteins with PSORT II [33] predicts the nuclear chloroquine, a lysosomal disrupting agent, enhanced the
localization scores of the TAT-Mu, Mu and Mu-Mu to be reporter gene expression significantly (data not reported)
similar. The contribution of arginines as a key feature in indicating the involvement of endocytotic pathways in
nuclear membrane translocation and localization has been the uptake of DPL. These results demonstrate that the
reported previously [34,35]. Recently, Miller’s laboratory internalization of the DNA is enhanced in the presence
has shown that Mu functions as an NLS, since a fusion of peptide. Although we observed clear enhancements
of Mu with β-galactosidase was shown to efficiently in transfection efficiencies with DP complexes, the exact
accumulate in the nucleus [36]. This is also supported contributions of TAT-Mu, Mu and Mu-Mu in enhancing
by our results where all the three fusion proteins have DNA delivery needs to be investigated. Nevertheless, the
been shown to internalize efficiently and localize in the observed uptake, co-localization and immunolocalization
nucleus thereby transfecting various cell lines. data when combined would explain the efficiency of
TAT is probably the most investigated among PTDs and transfection observed by us.
extensive literature is available on its ability to deliver Cationic peptides, although less commonly used, are
proteins and genes into cells [37–39]. The domain that however gaining attention due to their versatility and
confers this unique property to TAT is the stretch of advantages. By combining cues from viral proteins
YGRKKRRQRRR, rich in arginine and lysine, responsible with that of current transfection protocols that employ
for membrane transduction and also nuclear localization cationic lipids, novelty combined with multifunctionality
[38]. Mu-Mu alone was more efficient than Tat-Mu is introduced making them more stable, potential, next-
or Mu in binding with the DNA as seen from the generation formulations. Cationic lipids do not offer all
DNA-binding assays and EtBr-exclusion experiments. The the features that are required for a unique delivery system
binding ability of Mu is also reflected in its ability to and most current methods seek to generate formulations
bring about transfection. Reporter gene expressions with not confined to cationic lipids alone. The benefit of using
the three peptides indicated that expression levels are recombinant peptides is the versatility that can be added
more or less comparable and correlate with the increased to the recombinant molecule by genetic engineering
uptake as seen from the confocal experiments. Although methods to suit various cell types. The addition of cell-
all the three peptides show efficient internalization of specific epitopes would make these heterologous fusions
plasmid DNA, these have been demonstrated to be more therapeutically significant particularly in differentiated
efficient when combined with cationic lipids. Peptides cells as NLS-like features characterize these constructs.
when combined with lipids have been shown to be more This would also advance the development of vectors
efficient in bringing about reporter gene expression than where a combination vector system could be reconstituted
peptides alone. depending on desired protocol.
For more than a decade, linear poly-l-lysine has
been used in gene delivery. Modifications or variations
in the length of the positively charged polymeric
scaffold can have a varying effect on transfection Conclusions
efficiency thereby contributing to the development of
the oligolysine molecules also as gene delivery agents. Recombinant proteins that are heterologous fusions with
Several studies have employed poly-l-lysine of different DNA binding and nuclear localization epitopes of viral ori-
sizes for binding and condensation. These complexes are gin, generated in this study, have considerable potential
effective at concentrations which are usually toxic to to facilitate DNA delivery and enhance transfection. By
cells [40–42]. Although poly-l-lysine served as a good using this strategy with cues from viral proteins, versatility

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
Recombinant Fusion Proteins Mediate Efficient Gene Delivery 285

to the molecule can be added to the modules rapidly in a 12. Loyter A, Scangos GA, Ruddle FH. Mechanisms of DNA uptake
by mammalian cells: fate of exogenously added DNA monitored
combinatorial fashion. Unique cell-targeting moieties can
by the use of fluorescent dyes. Proc Natl Acad Sci U S A 1982;
be added to these basic constructs thereby making them a 79: 422–426.
more specific-potential transfecting agent to suit all gene 13. Murray KD, Etheridge CJ, Shah SI, et al. Enhanced cationic
liposome-mediated transfection using the DNA-binding peptide
delivery strategies. An important aspect that emerges from mu (mu) from the adenovirus core. Gene Ther 2001; 8: 453–460.
this study is that although the recombinant proteins have 14. Sorgi FL, Bhattacharya S, Huang L. Protamine sulfate enhances
been shown to efficiently transfect actively dividing cells, lipid-mediated gene transfer. Gene Ther 1997; 4: 961–968.
15. Sloots A, Wels WS. Recombinant derivatives of the human high-
the presence of NLS domains in these fusion proteins may mobility group protein HMGB2 mediate efficient nonviral gene
confer the potential to transfect non-dividing/resting cells. delivery. FEBS J 2005; 272: 4221–4236.
This would impact strategies using ex vivo cell therapies. 16. Demirhan I, Hasselmayer O, Chandra A, Ehemann M, Chan-
dra P. Histone-mediated transfer and expression of the HIV-1
In situations where the nuclear membrane dissolution tat gene in Jurkat cells. J Hum Virol 1998; 1: 430–440.
does not take place, the complexes could gain entry into 17. Balicki D, Reisfeld RA, Pertl U, Beutler E, Lode HN. Histone
the nucleus and subsequently become available to the H2A-mediated transient cytokine gene delivery induces efficient
antitumor responses in murine neuroblastoma. Proc Natl Acad
transcriptional machinery. This would have tremendous Sci U S A 2000; 97: 11500–11504.
therapeutic significance and would advance studies in the 18. Li S, Huang L. In vivo gene transfer via intravenous
direction of gene medicine. administration of cationic lipid-protamine-DNA (LPD)
complexes. Gene Ther 1997; 4: 891–900.
19. Hyndman L, Lemoine JL, Huang L, Porteous DJ, Boyd AC,
Nan X. HIV-1 Tat protein transduction domain peptide facilitates
Acknowledgements gene transfer in combination with cationic liposomes. J Control
Release 2004; 99: 435–444.
The authors would like to thank the Department of Biotechnology 20. Akita H, Tanimoto M, Masuda T, et al. Evaluation of the
for a DBT fellowship to RR. We would also like to thank nuclear delivery and intra-nuclear transcription of plasmid DNA
Prof. David Dean, Northwestern University, Chicago, for going condensed with micro (mu) and NLS-micro by cytoplasmic and
nuclear microinjection: a comparative study with poly-L-lysine.
through the initial draft of the manuscript. We acknowledge J Gene Med 2006; 8: 198–206.
Soumya Sudhakar, summer student, in the preparation of the 21. Anderson CW, Young ME, Flint SJ. Characterization of the
pMu construct. adenovirus 2 virion protein, mu. Virology 1989; 172: 506–512.
22. Hosokawa K, Sung MT. Isolation and characterization of an
extremely basic protein from adenovirus type 5. J Virol 1976;
17: 924–934.
Supplementary Material 23. Varga CM, Wickham TJ, Lauffenburger DA. Receptor-mediated
targeting of gene delivery vectors: insights from molecular
mechanisms for improved vehicle design. Biotechnol Bioeng
The supplementary electronic material for this paper 2000; 70: 593–605.
24. Elouahabi A, Ruysschaert JM. Formation and intracellular
is available in Wiley InterScience at: http://www. trafficking of lipoplexes and polyplexes. Mol Ther 2005; 11:
interscience.wiley.com/jpages/1099-498X/suppmat/. 336–347.
25. Wadia JS, Dowdy SF. Modulation of cellular function by TAT
mediated transduction of full length proteins. Curr Protein Pept
Sci 2003; 4: 97–104.
References 26. Nagahara H, Vocero-Akbani AM, Snyder EL, et al. Transduction
of full-length TAT fusion proteins into mammalian cells: TAT-
p27Kip1 induces cell migration. Nat Med 1998; 4: 1449–1452.
1. Felgner PL, Gadek TR, Holm M, et al. Lipofection: a highly 27. Becker-Hapak M, McAllister SS, Dowdy SF. TAT-mediated
efficient, lipid-mediated DNA-transfection procedure. Proc Natl protein transduction into mammalian cells. Methods 2001; 24:
Acad Sci U S A 1987; 84: 7413–7417. 247–256.
2. Miller AD. Cationic liposomes for gene therapy. Angew Chem Int 28. Kichler A, Leborgne C, März J, Danos O, Bechinger B. Histidine-
Ed Engl 1998; 37: 1768–1785. rich amphipathic peptide antibiotics promote efficient delivery
3. Lollo CP, Banaszczyk MG, Mullen PM, et al. Poly-L-lysine-based of DNA into mammalian cells. Proc Natl Acad Sci U S A 2003;
gene delivery systems. Synthesis, purification, and application. 100: 1564–1568.
Methods Mol Med 2002; 69: 1–13. 29. Gao X, Huang L. Potentiation of cationic liposome-mediated
4. Zaric V, Weltin D, Erbacher P, Remy JS, Behr JP, Stephan D. gene delivery by polycations. Biochemistry 1996; 35:
Effective polyethylenimine-mediated gene transfer into human 1027–1036.
endothelial cells. J Gene Med 2004; 6: 176–184. 30. Zaitsev SV, Haberland A, Otto A, Vorob’ev VI, Haller H,
5. Herz J, Gerard RD. Adenovirus-mediated transfer of low Bottger M. H1 and HMG17 extracted from calf thymus nuclei
density lipoprotein receptor gene acutely accelerates cholesterol are efficient DNA carriers in gene transfer. Gene Ther 1997; 4:
clearance in normal mice. Proc Natl Acad Sci U S A 1993; 90: 586–592.
2812–2816. 31. Balicki D, Beutler E. Histone H2A significantly enhances in vitro
6. Simon RH, Engelhardt JF, Yang Y, et al. Adenovirus-mediated DNA transfection. Mol Med 1997; 3: 782–787.
transfer of the CFTR gene to lung of nonhuman primates: 32. Hatefi A, Megeed Z, Ghandehari H. Recombinant polymer-
toxicity study. Hum Gene Ther 1993; 4: 771–780. protein fusion: a promising approach towards efficient and
7. Ali M, Lemoine NR, Ring CJ. The use of DNA viruses as vectors targeted gene delivery. J Gene Med 2006; 8: 468–476.
for gene therapy. Gene Ther 1994; 1: 367–384. 33. Paul Horton, K.-J.P.T.O.&.K.N. Protein Subcellular Localization
8. Kostarelos K, Miller AD. Synthetic, self-assembly ABCD Prediction with WoLF PSORT. Proceedings of the 4th Annual
nanoparticles; a structural paradigm for viable synthetic non- Asia Pacific Bioinformatics Conference APBC06 39–48, 2006;
viral vectors. Chem Soc Rev 2005; 34: 970–994. Available: http://wolfpsort.seq.cbrc.jp.
9. Zhou R, Geiger RC, Dean DA. Intracellular trafficking of nucleic 34. Futaki S. Arginine-rich peptides: potential for intracellular
acids. Expert Opin Drug Deliv 2004; 1: 127–140. delivery of macromolecules and the mystery of the translocation
10. Medina-Kauwe LK, Xie J, Hamm-Alvarez S. Intracellular mechanisms. Int J Pharm 2002; 245: 1–7.
trafficking of nonviral vectors. Gene Ther 2005; 12: 1734–1751. 35. Siprashvili Z, Scholl FA, Oliver SF, et al. Gene transfer
11. Graham FL, van der Eb AJ. A new technique for the assay of via reversible plasmid condensation with cysteine-flanked,
infectivity of human adenovirus 5 DNA. Virology 1973; 52: internally spaced arginine-rich peptides. Hum Gene Ther 2003;
456–467. 14: 1225–1233.

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
286 R. Rajagopalan et al.

36. Keller M, Harbottle RP, Perouzel E, et al. Nuclear localisation 40. Wu GY, Wu CH. Receptor-mediated in vitro gene transformation
sequence templated nonviral gene delivery vectors: investigation b y a soluble DNA carrier system. J Biol Chem 1987; 262:
of intracellular trafficking events of LMD and LD vector systems. 4429–4432.
Chembiochem 2003; 4: 286–298. 41. Wagner E, Zenke M, Cotton M, Beug H, Birnstiel ML.
37. Frankel AD, Pabo CO. Cellular uptake of the tat protein from Transferrin-polycation conjugates as carriers for DNA uptake
human immunodeficiency virus. Cell 1988; 55: 1189–1193. into cells. Proc Natl Acad Sci U S A 1990; 87: 3410–3414.
38. Schwarze SR, Ho A, Vocero-Akbani A, Dowdy SF. In vivo protein 42. Kircheis R, Wagner E. Polycation/DNA complexes for in vivo
transduction: delivery of a biologically active protein into the gene delivery. Gene Ther Regul 2000; 1: 95–114.
mouse. Science 1999; 285: 1569–1572.
39. Jeang KT, Xiao H, Rich EA. Multifaceted activities of the HIV-
1 transactivator of transcription, Tat. J Biol Chem 1999; 274:
28837–28840.

Copyright  2007 John Wiley & Sons, Ltd. J Gene Med 2007; 9: 275–286.
DOI: 10.1002/jgm
Supplementary information for the online version
of the paper ‘Recombinant fusion proteins TAT-Mu,
Mu and Mu-Mu mediate efficient non-viral gene
delivery’

Figure S1.

You might also like