Gala Szoka

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Advanced Drug Delivery Reviews 56 (2004) 967 – 985

www.elsevier.com/locate/addr

GALA: a designed synthetic pH-responsive amphipathic peptide


with applications in drug and gene delivery
Weijun Li a, Francßois Nicol a,b, Francis C. Szoka Jr. a,*
a
Department of Biopharmaceutical Sciences and Pharmaceutical Chemistry, School of Pharmacy,
University of California at San Francisco, San Francisco, CA 94143-0446, USA
b
ALZA Corporation, 1900 Charleston Road, Mountain View, CA 94039-7210, USA
Received 16 July 2003; accepted 31 October 2003

Abstract

GALA is a 30 amino acid synthetic peptide with a glutamic acid-alanine-leucine-alanine (EALA) repeat that also
contains a histidine and tryptophan residue as spectroscopic probes. It was designed to explore how viral fusion protein
sequences interact with membranes. The sequence selected was long enough to span a bilayer in the a-helix, the
glutamic acids (Glu) were selected to provide a pH-dependent negatively charged side-chain and the EALA repeat was
adjusted so that the peptide would have a hydrophobic face of sufficient hydrophobicity to interact with the bilayer when
the peptide was in an a-helix. GALA converts from a random coil to an amphipathic a-helix when the pH is reduced
from 7.0 to 5.0. At neutral pH, GALA is water soluble while at acid pH, GALA binds to bilayer membranes. The
nature of the association and the type of peptide – peptide interactions in the membrane depend upon the physico-
chemical properties of the bilayer such as the acyl chain composition of the phospholipids and the presence of
cholesterol. Neutral and negatively charged bilayers composed of saturated phospholipids of 14 – 16 acyl chain length are
solubilized into peptide – lipid discs by GALA. GALA can induce fusion between small unilamellar vesicles (SUV)
composed of unsaturated phospholipids. Most importantly GALA forms a transmembrane peptide pore comprised of
approximately 10 GALA a-helical monomers that are arrayed in an a-helix perpendicular to the plane of the membrane.
Membrane leakage from neutral or negatively charged vesicles at pH 5.0 can be adequately explained by a mathematical
model assuming that GALA becomes incorporated into the vesicle bilayer and aggregates to form a transbilayer pore
consisting of 10 ( F 2) peptides. The lipid compositions of model bilayer have important effects on the GALA
transbilayer insertion mechanism and peptide orientation. Insertion of the pore into the membrane dramatically accelerates
transmembrane phospholipid flip-flop. Cationic peptides designed based upon GALA but containing a lysine-alanine-
leucine-alanine (KALA) motif can interact with nucleic acids and perturb biomembranes. The pH-controlled membrane

Abbreviations: ANTS, 1-aminonaphthalene-3,6,8-trisulfonic acid; ATR, infrared attenuated total-reflection spectroscopy; BODIPY,
dipyrrometheneboron difluoride; CD, circular dichroism; DE, 1,2-dielaidoyl; DM, 1,2-dimyristoyl; DO, 1,2-dioleoyl; DP, 1,2-dipalmitoyl; DPe,
1,2-dipetroselinoyl; DPX, p-xylene-bis-pyridinium bromide; FITC, fluorescein isothiocyanate; FTIR, fourier transform infrared spectroscopy;
HA, hemagglutinin; LUV, large unilamellar vesicle; NBD, N-4-nitrobenzo-2-oxa-1,3-diazole; ODN, oligonucleotide; PC, phosphatidylcholine;
PE, phosphatidylethanolamine; PEG, poly(ethylene glycol); PEI, polyethylenimine; PG, phosphatidylglycerol; PO, 1-palmitoyl-2-oleoyl;
Poly(DMAEMA-NVP), poly(2-(dimethylamino)ethyl methacrylate-co-N-vinyl-2-pyrrolidone); Rh, rhodamine; SUV, small unilamellar vesicle.
* Corresponding author. Tel.: +1-415-476-3895; fax: +1-415-476-0688.
E-mail address: szoka@cgl.ucsf.edu (F.C. Szoka).

0169-409X/$ - see front matter D 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2003.10.041
968 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

permealization induced by GALA and related peptides serve as a paradigm for the design of environmentally responsive
peptidic delivery vehicles for drugs and genes.
D 2004 Elsevier B.V. All rights reserved.

Keywords: GALA; KALA; Amphipathic peptide; Pore formation; Bilayer; Gene delivery; pH-sensitivity

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 968
2. Peptide mimic of the natural membrane active elements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 969
2.1. Natural amphipathic a-helical peptide, ion channel motif and viral fusion protein . . . . . . . . . . . . . . . . . . 969
2.2. GALA and related peptides: simplified synthetic amphipathic a-helices. . . . . . . . . . . . . . . . . . . . . . . 970
3. Pore formation mechanism of GALA and related peptides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
3.1. GALA properties in aqueous solution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
3.2. GALA interactions with model lipid membranes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 973
3.2.1. pH-dependent GALA binding to vesicles made of unsaturated PC lipids . . . . . . . . . . . . . . . . . . 973
3.2.2. GALA induces the fusion of SUV made of unsaturated PC lipids at low pH . . . . . . . . . . . . . . . . 974
3.2.3. GALA induces the fragmentation of LUV made of saturated PC lipids at neutral pH . . . . . . . . . . . . 975
3.2.4. pH-dependent pore formation by GALA in LUV . . . . . . . . . . . . . . . . . . . . . . . . . . . . 975
3.2.5. Peptide orientation on membrane . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 978
4. Applications for the delivery of plasmid DNA and oligonucleotide (ODN) . . . . . . . . . . . . . . . . . . . . . . . . . 980
5. What are the advantages of GALA-like peptides as components of a drug and gene delivery system? . . . . . . . . . . . . 982
6. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 982
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 983
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 983

1. Introduction 16 years exploring the details concerning how mem-


brane composition influences the strength of the
Biological membranes are the barriers that separate GALA –lipid interaction as well as the nature of the
the machinery of life from the chaotic environment. In resulting peptide structure in the bilayer [1– 10]. This
mammalian cells membranes are predominantly com- information is the basis of more recent attempts to
posed of proteins and amphipathic lipids: sphingoli- design peptides that can be applied for the delivery of
pids, phospholipids and cholesterol. The lipids genes and other nucleic acid drugs [11– 13]. Such
spontaneously assemble into a bilayer which is a bioresponsive synthetic peptides have the potential to
two dimensional solvent for the membrane proteins. be the basis of a highly efficient nucleic acid delivery
It has been postulated that the self-organizing ability system.
of lipids into bilayer vesicles was one of the earliest Rules for the design of peptide-based gene delivery
steps in the development of life. Indeed, an intact systems must take into account DNA condensation,
membrane is critical for proper cell function but it is active targeting to the biological membrane, efficient
also one of the major impediments for the delivery of transmembrane transport and nucleus access [14].
therapeutic agents into cells. This is particularly true GALA and GALA-like peptides are concerned with
for macromolecular drugs such as oligonucleotide the regulated permeabilization of membranes. Natural
(ODN) and DNA. Viruses have evolved proteins to pore-forming peptide, ion channel motif and viral
breach membranes, so we originally designed and fusion proteins provide classic examples on the natural
synthesized GALA to serve as a simple model of a process of peptide-induced membrane permeabiliza-
viral fusion peptide sequence. We believed GALA tion [15,16]. With the knowledge on the amphipathic
would aid in the understanding of how an amphipathic peptide– membrane interaction, we designed a series
sequence can perturb bilayers and induce fusion of amphipathic peptide vectors for the delivery of
between membranes [1,2]. We have spent the past gene-coded plasmid DNA or ODN [11,12,17,18]. In
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 969

this review, we will discuss the factors that influence fashion and induce the leakage of molecules pre-
pore-forming phenomena as revealed in the mecha- encapsulated in liposomes. In the most common
nisms of GALA and related amphipathic peptides, and model proposed to explain the amphipathic a-helices
suggest the ways they might be applied for gene permeabilizing lipid bilayer, an aqueous pore is
delivery. generated in the membrane as a result of the
oligomerization of membrane-bound peptides. These
ion pores are generally described as bundles of
2. Peptide mimic of the natural membrane active aligned transbilayer helices, with the hydrophilic
elements side of the helices forming the water-exposed inner
surface of the pore and the hydrophobic side
2.1. Natural amphipathic a-helical peptide, ion interacting with the fatty acyl chains of the lipids
channel motif and viral fusion protein [26 – 28].
Based on their potential to form aqueous pores in
The amphipathic helix is a peptide motif in which model lipid membranes, amphipathic a-helical pep-
the amino acids are arrayed in an a-helix with one tides are relevant models of ion channel proteins
face, composed of hydrophilic residues (mainly com- [29,30]. Ionic channels generally are large, complex
posed of polar/charged amino acids) while the oppo- multimeric transmembrane glycoproteins that facili-
site face is hydrophobic (composed primarily of tate the selective translocation of ions across lipid
apolar amino acids) [19]. The amphipathic helix is a bilayer. For example, a basic structural motif of the
widespread feature in many biologically active pep- nicotinic acetylcholine receptor superfamily is con-
tides and proteins of diverse origin and function [20 – stituted by five parallel transbilayer amphipathic a-
23]. The amphipathicity enables the a-helix to form helices in the central pore of the ion channel [31,32].
an interface between the apolar region of macromo- Bundles of transbilayer helices are thought to occur
lecules and the polar environment. In particular, also in a wide range of transport proteins such as
amphipathic a-helices can self-associate in water by bacteriorhodopsin [33], the sugar transporter lactose
hydrophobic interactions between apolar side chains permease [34] and ‘‘ABC’’-transport proteins [35].
and hence can control protein folding pathways or Amphipathic a-helical toxins that are able to asso-
anchor tertiary and quaternary protein structures. They ciate in membranes to form ion channels are valu-
are also able to associate with the hydrophobic regions able models for studying the properties of the pore
of a lipid bilayer. formed by these proteins [27]. This is particularly the
This ability to interact with interfaces is a double case because the pore comprises only one type of
edge sword and numerous organisms secrete short helix, which simplifies its structure. The study of
cytotoxic peptides that adopt an amphipathic a- these peptides, therefore, increases our understanding
helical conformation when they are bound to lipid of certain biological processes, such as nerve con-
membranes. In turn there are a spectrum of cells that duction, mass and information transport, energy
are sensitive to the cytotoxic action of these pep- conversion and cellular signaling [36].
tides, from bacteria, viruses and fungi to tumor and Membrane fusion proteins play important roles in
mammalian cells [24,25]. The cytotoxic peptides kill the process of viral entry into living cells. Most
the target cells by inducing permeabilization or viruses have a single integral fusion protein which
disruption of the plasma membrane [21]. It is now can bind to the cell membrane upon a conformation-
generally accepted that many of these peptides exert al change by a physiological trigger such as the pH-
their cytotoxic activities directly on the lipid matrix drop in endosome, binding to a receptor, or proteol-
of the cell membrane. These natural amphipathic a- ysis which facilitate the viral membrane fusion with
helical toxins induce drastic changes in the structure, the target cell membrane [37]. The fusion peptide
dynamics and properties of the lipid bilayers they usually presents on the N-terminal hydrophobic re-
interact with. In particular, they permeabilize model gion of transmembrane proteins of various viruses,
lipid membranes, i.e. they generate ion conduction such as retroviruses, orthomyxoviruses and para-
across planar lipid bilayers in a voltage-independent myxoviruses [38]. The viral fusion process can be
970 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

cataloged into two classes, low pH-dependent or pH- acronym (glutamic acid-alanine-leucine-alanine) for a
independent. membrane destabilizing peptide than was EALA so
The influenza virus hemagglutinin (HA) has been vanity won out over science in the naming of this
extensively investigated as a model of membrane peptide sequence.
fusion protein. To infect the host cell, the influenza GALA is a synthetic peptide designed to interact
virus binds to the cellular membrane first, and then with lipid bilayer preferentially at acidic pH [1]
internalized into the endosome. The low pH in the (Fig. 1). The sequence of GALA is (WEAALAEA-
endosome induces a loop – helix conformational LAEALAEHLAEALAEALEALAA). The factors
change in HA, leading to the insertion of a hydro- considered for choosing the peptide sequence were
phobic fusion peptide domain (HA-2) into both the (a) hydrophobicity and conformational preference of
viral and host membranes. This induces the two the residues, (b) length of the peptide, (c) type of pH-
adjacent membranes to form a ‘‘stalk’’ intermediate titratable amino acids, and (d) topology of the residues
structure, followed by a complete fusion of the two on the peptide when set in an a-helix. The sequence
membranes [37]. The viral genome is subsequently was chosen to increase the peptide hydrophobicity,
released into the cytosol. Contrasted to this, the HIV and therefore its interaction with bilayers, at low pH.
fusion protein (gp41) is pH-independent, inducing In addition, amino acids with a high a-helical pro-
membrane fusion at neutral pH. The ability of pensity were selected. Apolar leucine residues were
synthetic gp41 fusion peptides to perturb and disrupt introduced to confer a relatively high hydrophobicity
bilayer has been demonstrated to be dependent on on the peptide, whereas polar glutamic acid (Glu)
the peptide sequence and temperature [39]. It should residues were included to add a pH-dependent com-
be noted that the research on the interaction between
peptide mimics of viral fusion proteins and mem-
branes has both theoretical and industrial importance.
For example, the virosomes, the particles of mem-
brane (liposome) reconstituted with viral envelope
proteins such as the fusion protein, have been
developed as a vaccine with enhanced immune
sensitivity [40].

2.2. GALA and related peptides: simplified synthetic


amphipathic a-helices

Generally, the structure – activity relationship for


naturally occurring membrane active peptides is dif-
ficult to replicate [26,41]. To investigate more specif-
ically the exact role of peptide hydrophobicity, net
charge, amphipathicity and helicity in the membrane-
perturbing activities of amphipathic a-helices, syn-
thetic peptides with highly simplified sequences have
been studied. These peptides generally contain four or
fewer types of amino acids that are selected for their
helix-forming capabilities, as well as their polar/ Fig. 1. Structure representation of the GALA (up) and KALA
charged or apolar character. Our group has extensively (down) peptide [1,12]. The GALA peptide sequence is: WEAA-
investigated a synthetic amphipathic a-helix, desig- LAEALAEALAEHLAEALAEALEALAA; KALA sequence is:
nated GALA, that perturbs model lipid membranes WEAKLAKALAKALAKHLAKALAKALKACEA. The atoms of
oxygen, carbon, nitrogen, hydrogen and sulfur are represented by
and forms aqueous pore [1 –5,7 – 9]. GALA is a mis- red, light blue, dark blue, white and yellow color. Right parts are
nomer for this sequence since the repeat unit is actual- face-view of peptide C-terminals and the left are features along the
ly EALA, however GALA was considered a jazzier peptide helix axis.
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 971

ponent to the hydrophobicity and net charge. A 3. Pore formation mechanism of GALA and
sequence composed of 30 amino acids was chosen related peptides
to yield a stable a-helix at low pH that was long
enough to cross a bilayer. GALA can be essentially 3.1. GALA properties in aqueous solution
described as a repeat of the sequence glutamic acid-
alanine-leucine-alanine. Deviations from this repeat In aqueous solution, GALA was designed to be
were made in the peptide sequence to localize the unordered at neutral pH and a-helical below pH 6.0
hydrophobic leucine residues on one side of the helix [1,4,43]. Circular dichroism (CD) measurements
and the pH-titratable hydrophilic Glu residues on the demonstrated the induction of a a-helical conforma-
other side. Alanine residues were selected as spacers tion as the pH was decreased from 7.5 to 5.0. The a-
between the polar and the apolar faces of the helix. At helical content at pH 7.5 in a buffer containing 100
neutral pH, electrostatic repulsions between the car- mM KCl was 7% and independent of peptide con-
boxylic acid moieties of the Glus are expected to centration below 16 AM. Above this concentration, it
destabilize the helix, whereas at pH 5.0 the neutrali- increased and reached 23% at 74 AM, indicative of
zation of these groups should promote helix forma- GALA self-association [4]. For a peptide concentra-
tion. Moreover, when the Glu residues are protonated tion of 16 AM, the percentage of a-helical structure
the hydrophobicity of the Glu side chain increases increased when lowering the pH from 7.5 to 3.6,
[42]. The spatial segregation of the polar and apolar reaching a maximal value of 36% at pH 5.0 in a
amino acids on opposite sides of the helix confers a sodium acetate buffer [1]. The pH at the midpoint of
pronounced amphipathicity on the peptide. Trypto- maximum change of helical content was 6.0, which
phan and histidine residues were introduced as fluo- is close to the pKa of Glu in polyglutamic acid. This
rescent and NMR probes, respectively. The fact that indicates that the change in a-helical content was
GALA was designed from simple principles makes correlated with the protonation of the Glu side
this peptide a good model to study the interplay of chains. At neutral pH, the alignment of the negative
components in the progression from choosing an charges on the peptide indeed prevents stabilization
amino acid sequence to its secondary structure and of the a-helix. The helix was found stable at pH 5.0
membrane-perturbing capabilities. from 10 to 40 jC and could be denatured only above
Based on the biophysical study of GALA peptide, 40 jC. In addition, the a-helical content of GALA
GALA together with other synthetic amphipathic conformation was sensitive to the ionic strength. At
peptides have been developed as vectors for DNA pH 6.5, it increased linearly upon increasing the salt
or ODN delivery. However, GALA itself can not concentration from 50 to 400 mM KCl. This effect
bind to DNA or ODN directly for the strong nega- was attributed to the screening of the carboxylates by
tive charges on both GALA and DNA backbone the cations [1].
repelling each other. KALA, an alternative choice to GALA secondary structure was also determined
GALA, is a synthetic cationic peptide, which can using the technique of infrared attenuated total-reflec-
bind directly to DNA or ODN [12]. The basic tion (ATR) spectroscopy, by analysis of the amide IV
concept behind the KALA sequence is to replace line shapes with Fourier self-deconvolution and curve
negatively charged Glu residue on GALA with fitting [43]. At pH 7.0, it was estimated that 19% of
positively charged lysine (Lys) in a similar position the amino-acids adopt an a-helical conformation,
on the peptide backbone while maintaining the other whereas 20% are organized in h-sheet structures. At
characteristics (Fig. 1). The strongly apolar and pH 5.0, the percentage of a-helix increased to 69%,
helix-forming Leu residue was to generate a peptide and h-sheet structure accounted for 28% of the
incorporated into the sequence of 30 amino acids conformation computed from fourier transform infra-
with several Leu-to-Lys repeated unit in the sequence red (FTIR) spectra. The discrepancies between CD [1]
(Fig. 1). The positive charges yield a high binding and FTIR techniques in estimating GALA secondary
affinity to the negatively charged nucleic acid, structure at pH 5.0 were explained by the light
whereas the hydrophobicity perturbs the lipid pack- scattering caused by peptide aggregation in the CD
ing of the membrane. measurements.
972 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

The carboxylic acid groups of the Glu residues are modifications were indicative of oligomer formation.
close enough to allow formation of divalent cation When the pH was rapidly changed from 7.5 to 5.0, a
bridges connecting adjacent groups. Interaction of drastic variation in Trp-1 fluorescence intensity (1.6
GALA with divalent cations at neutral pH should AM peptide) was observed, at the kmax found at pH
therefore result in charge neutralization and promote 5.0. This indicated that the environment of the tryp-
helix formation. At pH 7.5 the a-helical content tophan was altered in the 2-s mixing time of the
increased upon raising Ca2 + concentration, from 7% instrument. These results agree with CD studies [1]
without cation to 41% at 100 mM Ca2 + [1]. At a that demonstrated how GALA undergoes a reversible
concentration of 15 mM, Mg2 + was slightly more helix– coil transition as the pH is cycled between 5.0
efficient than Ca2 + in stabilizing the helix, while Zn2 + and 7.5. The influence of divalent cations was also
was as potent as Ca2 +. At pH 5.0, divalent cations investigated. In agreement with CD measurements,
only slightly increased the helicity, which confirmed the presence of 20 mM Ca2 + or Mg2 + at pH 7.5
that most of the carboxylic acids are neutralized at this induced a blue shift of kmax from 360 to 353 nm,
pH. whereas Zn2 + caused a further alteration to 344 nm.
GALA1, GALA2, GALA3 and GALA4 are short To evaluate the importance of the amino-acids
GALA analogues that were investigated to demonstrate topology in the GALA sequence, three amino acids
the importance of peptide length on the stabilization of were transposed. Leu was replaced by Glu at positions
its secondary structure. The sequence of these peptides 9, 17, and 25, and Leu replaced Glu at positions 11,
is (Glu-Ala-Leu-Ala)n, n = 1 –4, for GALA1 – 4, respec- 19, and 26. These transpositions resulted in a peptide
tively. The end effects are expected to dominate their designated LAGA, with little amphipathic character
conformation, allowing little stable secondary struc- when folded into an a-helix [4]. CD studies indicated
ture. They undergo, however, a change in hydropho- that LAGA was random coil at pH 7.5 whereas 22%
bicity when the carboxylic acid groups are protonated of the amino-acids adopted an a-helical conformation
at low pH. The CD spectra of GALA3 and GALA4 did at pH 5.0. The value observed at pH 5.0 was lower
not change when the pH of the solution was decreased than that of GALA, revealing a higher stability of the
from 7.5 to 5.0, showing that they were too short to amphipathic helix.
have any stable secondary structure [1]. However, the The internal motions of GALA and LAGA at pH
helicity of a peptide (named as SFP-1) composed 5.0 and 7.5 were studied over a broad range of peptide
essentially of the first 13 amino-acids of the GALA concentrations, by observing the anisotropy of the
sequence was shown by CD measurements to increase tryptophan moiety [4]. At pH 7.5, GALA and LAGA
from 3% at neutral pH to 41% at pH 4.6 [44]. Com- anisotropies (0.019 and 0.016, respectively) did not
pared to GALA3 or GALA4, the SFP-1 has an addi- change at concentrations lower than 10 AM. At 50 AM
tional Phe residue on the N-terminal. We believe that it increased to 0.142 for GALA, which was consistent
the hydrophobicity of Phe might help to stabilize the with the formation of aggregates at high peptide
helix at low pH. concentrations. The anisotropy of GALA at pH 5.0
The sensitivity of Trp-1 to the polarity of its was much higher than that observed at pH 7.5. At the
environment was also used to investigate the solution lowest concentration tested (0.33 AM), the value
properties of GALA. The fluorescence emission spec- obtained (0.046) was already significantly higher than
trum of GALA’s tryptophan (1.6 AM peptide) dis- that observed at pH 7.5. Increasing the peptide con-
played a maximum intensity that varied from centration at pH 5.0 resulted in a further increase in
kmax = 360 nm at pH 7.5 to kmax = 350 nm at pH 5.0 anisotropy up to 0.075 at 7 AM, which reflected
[1]. This blue shift indicated that the N-terminal changes in the self-association state of GALA. At this
tryptophan was in a more hydrophobic environment pH the anisotropy of LAGA (0.026) was much lower
at acidic pH, due to a modification of GALA’s and, contrary to GALA, was concentration-indepen-
conformation or to a different state of aggregation. dent. These results demonstrated that LAGA, because
A gradual blue shift of kmax, concomitant with an of the lack of amphipathicity of its helix, does not
increase in helicity, occurred at pH 7.5 upon increas- self-associate extensively at pH 5.0, whereas GALA
ing the peptide concentration above 20 AM [4]. These does and in a concentration-dependent manner.
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 973

Contrasted to GALA, the KALA peptide under- vesicles to the top of the gradient, while the unbound
goes a pH-dependent a-helix to random coil change fraction remained at the bottom. The apparent distri-
when pH is decreased from 7.5 to 5.0 [12]. The pH- bution coefficient (Ka=(bilayer-bound peptide/peptide
dependence of this peptide is based on the ionization in solution)  ([H2O]/[lipid])) in these conditions (2
properties of residues with charged side chains, in- AM peptide) was 2.0  105. This distribution coeffi-
cluding the q-amino group of seven Lys, the imidazole cient can also be considered as an apparent association
of histidine, and g-carboxyl groups of two Glu resi- constant. When incubated at pH 7.5 under similar
dues. When the pH is decreased from 8 to 4.5, the a- conditions, GALA remained largely at the bottom of
helix percentage in the peptide will decrease from the gradient, resulting in a very low apparent partition
45% to 24%. At pH 7– 8, KALA has five net positive coefficient (Ka = 1.8  103). This result reveals a dras-
charges ( + 7 from Lys residues and 2 from Glu), tic pH-dependence of GALA partitioning between
and the peptide keeps the a-helix. When the pH is lipid bilayers and water, and demonstrates the strong
decreased to 5, the charge on Glu residue will be propensity of the amphipathic helix to associate with
neutralized and the imidazole of histidine will be lipid vesicles.
protonated. The increased net charge (maximum + 8) When GALA was first incubated with egg PC
appears to disrupt the a-helix in aqueous solution liposomes at pH 5.0 and the pH was elevated to 7.5,
[12]. the results mimicked the outcome at pH 7.5, indicat-
ing that GALA dissociated from the vesicles as a
3.2. GALA interactions with model lipid membranes result of the pH elevation. Keeping the peptide con-
centration constant at 2 AM while lowering the lipid to
GALA’s ability to perturb lipid membranes has peptide ratio yielded an increase in GALA binding to
been extensively investigated. In particular, GALA egg PC vesicles at pH 5.0, from Ka = 2.0  105 at
was shown to induce membrane fusion [2], fragmen- 5000/1 to 4.5  106 at 50/1. An increase of the
tation [3], and permeabilization by pore formation apparent partition coefficient when the lipid concen-
[1,5]. In addition, the membrane-binding properties of tration is reduced indicates a positive cooperativity of
GALA have been studied to explain its ability to the binding process. This cooperativity can be attrib-
perturb lipid bilayers [1– 4,43]. In these studies GA- uted to GALA aggregation in egg PC bilayers at pH
LA was always added to the vesicles suspensions from 5.0.
a stock solution at pH>7.0, to avoid pre-aggregation When LAGA was mixed with egg PC liposomes at
of the peptide. Further investigations with fluores- pH 5.0 at a lipid/peptide ratio of 5000/1 the resulting
cence labeled-GALA revealed the orientation of pep- apparent association constant was 1.9  104, which is
tide in unilamellar vesicles [8,10]. The state of the an order of magnitude lower than that found for
peptide in membranes is influenced by the composi- GALA in similar conditions. Since the two peptides
tion of the membrane lipids, such as the charge, the have comparable average hydrophobicity and helical
length of acyl chain, the saturation or unsaturation of contents in solution at pH 5.0, the amphipathic helix
acyl chain, and the percentage of cholesterol [7,9]. and the self-assembly of GALA in membrane is the
reason for its bilayer association.
3.2.1. pH-dependent GALA binding to vesicles made The wavelength of tryptophan’s maximal emission,
of unsaturated PC lipids kmax, was also monitored to demonstrate an interac-
The apparent association of GALA and LAGA with tion between GALA and egg PC liposomes [1,4]. At
vesicles was measured at pH 5.0 and 7.5 from the pH 7.5 the kmax remained constant (360 nm) when the
separation of 125I-peptide – vesicle mixtures on discon- peptide (1.6 AM) was mixed with egg PC liposomes at
tinuous ficoll gradients [4]. When GALA and egg a lipid/peptide molar ratio varying from 50/1 to 500/1.
phosphatidylcholine (PC) large unilamellar vesicles At pH 5.0, the kmax shifted to 335 nm in the presence
(LUV) (diameter f100 nm) were mixed at a 5000/1 of egg PC vesicles, indicating an association with the
lipid to peptide molar ratio for 15 min at pH 5.0 and vesicles. The spectral changes observed upon lower-
then applied on a gradient prepared at the same pH, the ing the pH from 7.5 to 5.0 in the presence of egg PC
fraction of lipid-bound GALA co-migrated with the vesicles were reversed upon elevating the pH to 7.5.
974 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

The secondary structure of GALA when bound to due to the transfer of its energy to the acceptor
small unilamellar vesicles (SUV) (diameter f 50 nm) fluorophore Rh. Upon addition of peptide to induce
made of the lipid POPC at pH 5.0 was determined the fusion of labeled and unlabeled vesicles, lipid
using ATR spectroscopy, by analysis of the amide IV mixing resulted in a decrease of the bilayer concen-
line shapes with Fourier self-deconvolution and curve trations of the donor and acceptor fluorophores, which
fitting [43]. A single peptide conformation was reduced fluorescence energy transfer. The percentage
obtained at the two lipid/peptide molar ratios tested, of lipid mixing could be estimated from the degree of
i.e. 50/1 and 100/1, revealing a a-helical content of recovery of NBD fluorescence. When POPC SUV
100%. This result demonstrates that the amphipathic were used at a concentration of 100 AM, GALA-
helix is stabilized upon binding to POPC vesicles. The induced lipid mixing at pH 5.0 was significant only
lower values estimated from CD measurements with above a lipid to peptide molar ratio of 300/1 and was
egg PC LUV vesicles were explained by a lack of peptide concentration dependent, reaching a value of
correction for adsorption flattening, which is known 55% at a ratio of 50/1. In addition, the initial rate and
to cause a significant underestimation of the a-helical final extent of lipid mixing increased with the lipid
content of membrane proteins. Polarized FTIR experi- and peptide concentrations, at a constant lipid to
ments on oriented lipid multibilayers pre-formed with peptide ratio. This behavior suggests that vesicle
the peptide GALA indicated that the axis of the helix contact was required for lipid mixing. At pH 7.5 no
was essentially oriented parallel to that of the acyl lipid mixing was observed for any of the lipids
chains of the phospholipid, with the helical axis being studied, at the same lipid to peptide ratios of 50/1
possibly slightly tilted (10 – 15j) in respect to the axis and 100/1. This result confirms the drastic pH-depen-
of the hydrocarbon chains [43]. This result confirmed dence of GALA’s ability to perturb bilayers. Lipid
the interpretation by the tryptophan quenching experi- mixing does not result from vesicle lysis and refor-
ments that GALA exhibits a transverse orientation in mation, since a FITC-detran 4100 Mw pre-encapsu-
egg PC bilayers. As discussed later, GALA’s ability to lated in egg PC SUV remained entrapped after
permeabilize egg PC vesicles at pH 5.0 were attribut- addition of the peptide [2].
ed to the formation of aqueous pores composed of Lipid mixing at pH 5.0 was correlated with an
transmembrane helices. increase in light scattering, indicative of vesicle ag-
gregation or fusion [2]. Dynamic light scattering was
3.2.2. GALA induces the fusion of SUV made of used to obtain an average size of egg PC and POPC
unsaturated PC lipids at low pH vesicles before and after addition of GALA at a 50/1
GALA was originally designed to generate a water lipid to peptide ratio. Measurements demonstrated an
soluble pH-triggered peptide fusogen that can mimic increase of the vesicle size at pH 5.0, whereas no
the behavior of the fusogenic sequence of viral increase was observed at pH 7.5. In the case of egg PC
proteins. The influenza virus is internalized into endo- and POPC vesicles at pH 5.0, the size of the SUV
somes where the N-terminus of its HA protein is increased from 50 to 70 and from 50 to 100 nm,
rendered fusion-competent by a conformational respectively, after the addition of GALA [2]. When
change resulting from pH acidification [45,46]. Here, the pH was elevated to 7.5 after incubation of vesicles
the idea was to use the pH-dependence of both GALA with GALA at pH 5.0, the vesicles maintained their
conformation and its binding affinity to PC mem- size of 70 nm, which confirmed that the increase in
branes, to induce vesicle fusion preferentially at low lipid mixing at pH 5.0 was not due to reversible
pH. A lipid mixing assay was employed to monitor aggregation but fusion. In agreement with the fact
GALA-induced fusion of vesicles [2]. In this assay, a that fusion of POPC SUV terminated when a size of
population of vesicles containing 1 mol% of the two 100 nm was reached, LUV of 100 nm diameter did not
fluorescence labeled lipids, N-4-nitrobenzo-2-oxa-1,3- show any lipid mixing or size increase at pH 5.0 after
diazole (NBD)-phosphatidylethanolamine (PE) and GALA addition.
rhodamine (Rh)-PE was mixed in a 1:9 molar ratio A short peptide (SFP-3) corresponding to the 14
with probe-free vesicles. Before fusion of vesicles first amino-acids of the GALA sequence, with an
occurred, NBD fluorescence was partially quenched additional cysteine at the C-terminus, was conjugated
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 975

to egg PC/Cholesterol/SMCC-PE (6/3/0.5) liposomes measured by dynamic light scattering, was 30 nm.
to enhance the peptide’s ability to induce vesicles Negative stain electron microscopy of the complex
fusion [44]. The fusogenic potency of SFP-3 after revealed only stacked discoidal structures with an
coupling to 200 nm liposomes at a 400/1 lipid/peptide approximate edge thickness of 44 Å and diameters
molar ratio was studied using a lipid mixing assay between 170 and 350 Å. When the pH was subse-
based on the fluorescence energy transfer between quently lowered from 7.5 to 5.0, the apparent diameter
NBD and Rh-labeled lipids. No lipid mixing was of the GALA/DMPC complex increased to 415 nm
observed at pH 7.4 when mixing (SFP-3)-coupled [3]. On negative stain electron micrographs, exten-
Rh-labeled liposomes with (SFP-3)-coupled NBD- sively aggregated arrays of discoidal structures with
labeled liposomes nor at pH 4.5 when mixing an the same dimensions as those observed at pH 7.5 were
unconjugated peptide with Rh-labeled and NBD-la- present. However, when GALA was mixed with egg
beled liposomes. However, a fusion yield of 75% was PC LUV at pH 5.0 at a lipid/peptide ratio of 30/1, only
obtained at pH 4.5 when mixing (SFP-3)-coupled Rh- vesicle profiles could be observed [3].
labeled liposomes with (SFP-3)-coupled NBD-labeled Thus, GALA could interact with liposomes com-
liposomes. Light scattering measurements showed a posed of saturated phospholipids at neutral pH and
marked increase in the vesicle diameter from 235 nm form complexes similar to those described for apoli-
at pH 7.4 to 889 nm after 30 min at pH 4.5 [44]. poproteins [3]. CD measurements confirmed that
Compared to the unconjugated SFP-3, the lipid-an- GALA interacted with these vesicles at neutral pH.
chor of SFP-3-PE conjugate might facilitate the pep- Upon incubation of GALA with DMPC vesicles at 26
tide interaction with two adjacent bilayers and jC or with DPPC vesicles at 26 or 45 jC, an increase
promote membrane fusion by orienting the peptide of the peptide helicity was observed at a lipid/peptide
at the interface between the two membranes. ratio of 30/1 [3]. Using the technique of FTIR, a a-
helical content of 65% was determined at pH 7.0 with
3.2.3. GALA induces the fragmentation of LUV made DMPC vesicles [43]. At 26 jC, the increase in helical
of saturated PC lipids at neutral pH content with DMPC and DPPC vesicles was accom-
GALA’s interaction with vesicles made of the panied by a 7 nm blue shift of the tryptophan kmax,
unsaturated lipids at neutral pH is very weak [3]. while no shift was observed with DPPC at 45 jC.
Nevertheless, with liposomes made of the saturated In the DMPC/GALA discoidal structures formed at
lipids such as DMPC or DPPC, the situation is pH 7.5, GALA helix was oriented parallel to the lipid
drastically different [3]. Incubation of GALA (33 acyl chains, which supports the idea of a rim of
AM) with DMPC LUV at a lipid/peptide ratio of 30/ GALA surrounding a DMPC bilayer core [3]. The
1 resulted in a clarification of the liposome suspension same parallel orientation occurred when GALA inter-
within 30 min at 4 or 26 jC which is below or above acted at pH 5.0 with LUV composed of the unsatu-
the gel to liquid-crystalline transition temperature rated lipids POPC and egg PC, but in this case the
(Tm = 23 jC) of the lipid. A similar behavior was vesicle structure was retained. This behavior suggests
observed with DPPC vesicles, only when the mixture that the amphipathic a-helix forms transmembrane
was incubated a few degrees above the Tm of the lipid, aggregates, or aqueous pores, in the membrane of
i.e. 41 jC. In that case, the mixture remained optically LUV composed of unsaturated lipids. Aggregation of
clear when the temperature was decreased below the the discoidal DMPC/GALA particles upon lowering
Tm after a pre-incubation above the Tm. Surprisingly, the pH from 7.5 to 5.0 might reflect a transition
the negatively-charged GALA (pH 7.5) could also between these two states.
solubilize vesicles made of the saturated anionic lipid
DMPG. 3.2.4. pH-dependent pore formation by GALA in LUV
The GALA/DMPC complex was isolated by gel As described previously, GALA binding to ve-
filtration chromatography at pH 7.5. Although incu- sicles made of POPC or egg PC is highly pH-depen-
bated at a molar ratio of DMPC to GALA of 30/1, the dent. The binding is enhanced at low pH due to a
resulting complex had a stoichiometry of 19/1. The stabilization of GALA amphipathic a-helix. As dis-
apparent diameter of the GALA/DMPC complex, as cussed in the second part, it is believed that the ability
976 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

of natural amphipathic helical peptides to permeabi- was sufficient to induce 100% leakage. The enhanced
lize vesicles or cells is related to their propensity to GALA activity observed when the pH is decreased
adopt such a conformation. Therefore, GALA’s ability from 7.5 to 5.0 correlates with the increase in peptide
to induce the release of contents from LUV made of helicity and membrane-binding.
the lipid egg PC was investigated using an ANTS/ In all conditions tested, the leakage reached a
DPX leakage assay [1,5]. In this assay, 1-aminonaph- stable but different plateau of fluorescence value
thalene-3,6,8-trisulfonic acid (ANTS) and p-xylene- f 10 min after addition of the peptide to the lip-
bis-pyridinium bromide (DPX) are encapsulated in osomes [1,5]. If the peptide transferred among the
egg PC LUV at concentrations where DPX quenches vesicles, eventually all of the contents would be
ANTS fluorescence (Fig. 2). When a vesicle is per- released. Thus this analysis revealed that the leakage
meabilized, release of its contents in the external induced by GALA proceeded via an ‘‘all or none’’
milieu produces a dilution of ANTS and DPX, which mechanism, i.e. vesicles either leaked their entire
highly reduces the quenching. The extent of ANTS/ content in ANTS/DPX or did not leak at all. Thus,
DPX leakage can be estimated from the fluorescence the percent increase in ANTS fluorescence observed
increase. at the plateau corresponded to the percent of vesicles
GALA’s ability to induce ANTS/DPX leakage that had leaked their entire content in ANTS and
from egg PC LUV was highly dependent on the pH DPX. This behavior suggests that there were a critical
of the solution from 4.5 to 7.5 [1,5]. At each pH number of peptides that needed to be bound to a
tested, a decrease in the lipid to peptide molar ratio at vesicle to generate leakage of its contents, and the
a constant lipid concentration (0.1 mM), i.e. an peptide did not readily transfer among the vesicles at
increase in the peptide concentration, resulted in a low pH.
higher extent of leakage. At pH 4.5 and 5.0, GALA A model was proposed to describe GALA-in-
displayed similar behaviors and was considerably duced ANTS/DPX leakage at pH 5.0 [5,8] (Fig. 3).
active, since a lipid/peptide molar ratio of 2500/1 The model predicts that when GALA is added to a
vesicle suspension, the peptide binds rapidly to the
lipid membrane where it aggregates to form aqueous
pores. In a pore, GALA has a predominantly a-he-
lical secondary structure with the helix axis aligned
perpendicular to the bilayer surface, as confirmed by
FTIR measurements [5,10]. Once a pore is assem-
bled, the entire vesicle content is rapidly released in
the external milieu. It is possible that the channel
assembles by the stepwise addition of monomers or
through the association of multimeric aggregates. In
either case, leakage only occurs when the channel
becomes large enough (M monomers) to permit
diffusion of the entrapped molecules. The fact that
ANTS/DPX leakage proceeded by an ‘‘all or none’’
mechanism suggests that once a pore is formed,
release of vesicle contents is fast. In addition, the
rapid changes in tryptophan emission maximum
observed when the peptide is added to an egg PC
Fig. 2. Schematic kinetics of GALA-induced ANTS/DPX leakage liposome suspension at pH 5.0 confirms that GALA
assay at pH 5.0. In this assay, ANTS and DPX are encapsulated in binding to vesicles is a rapid process. A mathemat-
vesicle at concentrations where DPX quenches ANTS fluorescence. ical analysis of the percent of leakage obtained at
When a vesicle is permeabilized by GALA, release of its contents in
the external milieu produces a dilution of ANTS and DPX, which various lipid to GALA molar ratios at pH 5.0 was
highly reduces the quenching. The extent of ANTS/DPX leakage attempted. Since from the lipid and peptide concen-
can be estimated from percentage of the fluorescence increase. trations, the fraction of bound peptide at these
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 977

Fig. 3. Model of pore formation by GALA at acidic pH. The model predicts that when GALA is added to a vesicle suspension, the peptide binds
rapidly to the lipid membrane surface where it forms as planar aggregates parallel to the membrane surface with the apolar side of the helix
slightly embedded in the membrane and the polar side facing water. Along with the increase of peptide concentration in the membrane, the
planar aggregates expand and insert into the membrane to form aqueous pores. In a pore, GALA has a predominantly a-helical secondary
structure with the helix axis aligned perpendicular to the bilayer surface [5,10]. Once a pore is assembled, the entire content encapsulated in
vesicle is rapidly released to the external environment. It is possible that the channel assembles by the stepwise addition of monomers or through
the association of multimeric aggregates. In either case, content leakage only occurs when the channel becomes large enough (M monomers) to
permit diffusion of the entrapped molecules.

concentrations and the size distribution of the lip- 445 to 5200 Mw confirmed that 10 F 2 monomers for
osomes are known, the model can predict the distri- pore formation is consistent with the data [5]. At a
bution of the number of membrane-bound peptides lipid to peptide molar ratio of 2500/1, ANTS (445
per vesicle. The model assumes that each vesicle that Mw), NADH (660 Mw), and acid blue 9 (795 Mw)
contains i z M peptides bound to its bilayer will were released from the vesicles, whereas AP5A (915
contain a pore that allows leakage of ANTS/DPX Mw) and inulin (5200 Mw) were retained. The pore
(aggregates composed of i z M monomers), which diameter was estimated as a function of the number of
corresponds to a situation where GALA aggregation helices. Given the Stokes radius of the vesicle-encap-
in the membrane is irreversible at low pH such as sulated compounds, a pore of 8 – 12 monomers could
pH 5.0 (Fig. 3). The percent of leakage was pre- explain the retention of AP5A and inulin and the
dicted as a function of the lipid to peptide molar release of ANTS, NADH, and acid blue 9. The ability
ratio for several M values. Calculated values agreed of the model to explain and predict the kinetics of
with the experimental percents of leakage when the leakage was also tested. It was assumed that the rate-
minimum size of the pore that allows ANTS/DPX limiting step for ANTS/DPX leakage is the formation
leakage was set to M = 10 monomers. Taking into of an aggregate composed of M = 10 peptides in the
account the possible experimental errors on the lip- bilayer. When the forward rate constant of aggrega-
osomes size distribution and on the fraction of bound tion, was set to 0.002 s 1, and aggregate dissociation
peptide (0.645), the minimum M value was estimated was ignored, the calculated curves were in good
to be 10 F 2. The model has been improved to take agreement with the experimental kinetics, especially
into account the possibility of reversible surface within the first minute after peptide addition [5].
aggregation. Results establish that GALA aggrega- Using egg PC LUV asymmetrically labeled with
tion in POPC vesicles is quasi-irreversible at pH the fluorescent lipid probe NBD-PC, GALA was
5.0 [5,8]. shown to catalyze rapid translocation of the lipids
Leakage experiments with vesicle-entrapped com- from one bilayer leaflet to the other at pH 5.0 [6]. A
pounds of different molecular weights ranging from mathematical analysis of the kinetics and final
978 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

extents of NBD-PC flip-flop observed after addition DOPG. Results from binding experiments reveal that
of GALA to asymmetrically labeled vesicles was this reduced leakage from DOPC/DOPG vesicles
performed over a wide range of lipid to peptide cannot be explained by a reduced binding affinity
ratios (from 30/1 to 300,000/1). Two modes of of the peptide to these membranes. By monitoring
flip-flop were deconvoluted, one is related to pore the leakage of a fluorescent dextran, an increase in
formation and a second arising from peptide aggre- the minimum pore size also does not explain the
gates that are too small to form a pore. Analysis of reduction in ANTS/DPX leakage. The data suggest
the final extents of lipid flip-flop confirmed that that the bilayers composed of phospholipids contain-
GALA forms pores with a minimum size of ing a cis unsaturated bond per acyl chain (DO and
M = 10 F 2 monomers. In addition, the kinetic anal- DPe) can stabilize surface-associated GALA mono-
ysis predicted that the pore-mediated mode of flip- mers or aggregates, while the effect of peptide
flop proceeded with a rate constant kpore z 1 s 1, transbilayer insertion is reduced. GALA-induced
while the bilayer-perturbing mode was much slower ANTS/DPX leakage is also decreased when the
(kperturbation = 3  10 6). The estimated rate constant vesicles contain PE lipid with small hydrophilic head
of lipid flip-flop associated with pore formation group [9]. This lends further support to the pore-
corresponded to a situation where the asymmetry forming model that factors stabilizing the surface
of the probe distribution in the bilayer was relieved state of GALA reduce its insertion and subsequent
in less than 60 s after a pore had been formed. A pore formation in the bilayer (Fig. 4).
mechanism to account for the rapid flip-flop induced The effect of cholesterol on the pore formation
by GALA pore is that the phospholipid headgroup of GALA in LUV composed of neutral and nega-
resided in the water-filled pore, while the translocat- tively charged phospholipids was also determined
ing acyl chains remained within the hydrophobic [7]. Increasing cholesterol content in the membranes
interior of the bilayer. However, the peptide LAGA, resulted in a reduced efficiency of GALA to induce
which was unable to form discrete pores, catalyzed membrane leakage. Part of the cholesterol effect
flip-flop through bilayer perturbations, with a rate was due to reduced binding of the peptide to
constant comparable to that of the bilayer-perturbing cholesterol-containing membranes. An additional ef-
mode of GALA [6]. fect of cholesterol was to increase reversibility of
The above investigations on GALA induced pore surface aggregation of the peptide in the membrane.
formation were done using egg PC as membrane FRET experiments using NBD-labeled GALA and
model. To better understand the influence of phos- Rh-labeled GALA confirmed that the degree of
pholipid acyl chain composition on the pore forma- reversibility of surface aggregation of GALA was
tion by GALA, the ANTS/DPX leakage assay was significantly larger in cholesterol-containing lipo-
examined at pH 5.0 from LUV vesicles made of PC somes, thus reducing the efficiency of pore forma-
and phosphatidylglycerol (PG) with the following tion [7].
acyl chain compositions: 1-palmitoyl-2-oleoyl (PO),
1,2-dioleoyl (DO), 1,2-dielaidoyl (DE), and 1,2-dipe- 3.2.5. Peptide orientation on membrane
troselinoyl (Dpe) [9]. A mathematical model was In an attempt to resolve whether, at pH 5.0, the
able to predict and simulate the final extents of N-terminus of the membrane-bound GALA is pre-
GALA-mediated ANTS/DPX leakage assay from dominantly interacting with the bilayer surface or
POPC/POPG and DEPC/DEPG liposomes at pH whether it is embedded deeper in the bilayer core,
5.0 as a function of peptide concentration in the bromine-labeled lipids were employed as quenchers
bilayer, by considering that GALA pores responsible of tryptophan fluorescence [4]. The depth of pene-
for this leakage have a minimum size of 10 F 2 tration of the tryptophan within the bilayer can be
monomers and are formed by quasiirreversible ag- probed by including brominated lipids that position
gregation of the peptide [9]. With the phospholipid the bromine at different locations in the bilayer. For
acyl chain compositions tested, GALA-induced quenching to occur, bromine must contact or be
ANTS/DPX leakage follows the rank order POPC/ within 5 Å of the tryptophan moiety. 5,6-Dibromo-
POPG c DEPC/DEPG>DPePC/DPePG>DOPC/ cholesterol, which positions the bromine near the
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 979

Fig. 4. Reduced insertion of GALA aggregates with DOPC and DOPE. The bilayers composed of phospholipids containing a cis unsaturated
bond per acyl chain (DOPC) can stabilize surface-associated GALA monomers or aggregates, while the effect of peptide transbilayer insertion is
reduced. Peptide transbilayer insertion is also decreased when the vesicles contain PE lipid with small hydrophilic head group.

lipid headgroup region of the bilayer, and BrPC, will be enhanced [47]. BODIPY-avidin was either
which positions the bromide at the 9,10-position of added externally after vesicle formulation or was
the lipid acyl chains, were each incorporated into pre-encapsulated in vesicles to determine the fraction
egg PC LUV at varying mole percents. Both 10 of liposome-bound biotinylated GALA that exposed
mol% brominated cholesterol and 12 mol% BrPC its labeled terminus to the external or internal side of
were able to quench tryptophan fluorescence at pH the bilayer, respectively. Under conditions (at low
5.0 at lipid/peptide molar ratios of 50/1 (24% and lipid/peptide molar ratios such as 2500/1) where
31%, respectively) and 500/1 (31% and 33%, re- most of the membrane-bound peptides were involved
spectively). These extents of fluorescence quenching in transmembrane aggregates and formed aqueous
indicated that the N-terminus of GALA had pene- pores, the head-to-tail (N- to C-terminus) orientation
trated into the hydrophobic portion of the bilayer. It of GALA peptide was that 3/4 of the peptides
was proposed that GALA tryptophan is positioned exposed their N-terminus on the inside of the vesicle
closer to the headgroup – hydrocarbon interface than and their C-terminus on the outside, and 1/4 of the
to the interior of the bilayer, which is consistent peptides exposed their N-terminus outside and their
with a transbilayer orientation of GALA in egg PC C-terminus inside. Under conditions resulting in
liposomes. reduced amount of pore formation (at higher lipid/
We also determined the orientation of a biotiny- peptide molar ratios), an increase was found in the
lated version of the pore-forming peptide GALA at fraction of GALA termini exposed to the outside of
pH 5.0 in LUV by designing a dipyrrometheneboron the vesicle when biotinylated GALA was added
difluoride (BODIPY)-avidin fluorescence binding to externally. These results are consistent with the pore
a GALA biotinylated at the N- or C-terminus [8] formation model that requires a critical number of
(Fig. 5). The BODIPY-avidin/biotin binding assay peptides (M) in an aggregate to form a transbilayer
was developed based on the phenomenon that the structure [5]. When the aggregation size of GALA
green fluorescence intensity of a BODIPY-modified peptide is smaller than M, the peptide orientation is
avidin, upon its quasi-irreversible binding to D-biotin, mostly parallel to the membrane surface, and both N-
980 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

Fig. 5. GALA-termini targeting using BODIPY-avidin/biotin binding assay. The BODIPY-avidin/biotin binding assay was developed based on
the phenomenon that the green fluorescence intensity of a BODIPY-modified avidin, upon its quasi-irreversible binding to D-biotin, will be
enhanced [47]. BODIPY-avidin was either added externally after vesicle formulation or was pre-encapsulated in vesicles to determine the
fraction of liposome-bound biotinylated GALA that exposed its labeled terminus to the external or internal side of the bilayer, respectively. Both
N- and C-terminus of GALA can be biotinylated.

and C-terminus of the biotinylated peptide are ex- ery into the nucleus. Addition of a peptide that
posed to external BODIPY-avidin [8]. destabilizes membranes at low pH can mediate the
release of the DNA from the acidic endocytic com-
partment and enhance gene expression. In that respect,
4. Applications for the delivery of plasmid DNA synthetic pH-sensitive peptides derived from the N-
and oligonucleotide (ODN) terminal region of influenza virus HA subunit (HA2)
were shown to be particularly potent [50,51]. GALA
To a large extent, advances on gene therapy depend is considerably more active than these peptides in
on the efficient and safe design of gene delivery permeabilizing PC vesicles at low pH but when bound
vectors. Non-viral gene delivery systems are devel- by electrostatic interactions to a DNA/polylysine-
oped as a substitution for viral gene transfer vectors conjugated ligand (transferrin) complex, the GALA
because they may be safer and easier to manufacture. peptide was less efficient in enhancing gene expres-
Cationic systems that mimic aspects of the viral sion than the influenza-based peptides [51].
structure and its protein/membrane coating, can po- When DNA was complexed to a dendritic poly-
tentially protect the DNA from blood clearance, and amidoamine cationic polymer (dendrimer) to which
then transfer the gene into the targeting tissue. GALA was covalently attached to amines on the
The bilayer-destabilizing properties of GALA at dendrimer, an increased gene expression was ob-
low pH have been utilized to enhance the delivery of served [18]. This enhanced effect is an indicator that
genes into the nucleus of cells in vitro (Table 1). GALA motif can be covalent conjugated into suit-
Complexes prepared by mixing DNA with polylysine- able platform such as dendrimer to make an efficient
conjugated ligands can be efficiently delivered into gene transfer system. We observed that GALA when
cells by receptor-mediated endocytosis [48,49]. How- simply added to the dendrimer DNA complex, could
ever, such complexes accumulate in the endosome/ also improve gene transfer. We ascribe this to the
lysosome. This reduces the efficiency of DNA deliv- formation of a ternary complex between the compo-
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 981

Table 1
Formulation of DNA or ODN delivery systems based on GALA and related peptides
Fusogenic peptide Vector composition Reference
Nucleic acid condensing reagent Ligand
GALA Polylysine Transferrin [51]
GALA Cationic liposome (DOTAP, or DOTAP/DOPE) Transferrin [52 – 54]
GALA Dendrimer – [18]
EALA Cationic liposome (DOTAP/cholesterol/PEG-PE) Folate [66]
Gramicidin S Gramicidin Sa – [11]
KALA – – [12,67]
KALA PEI – [55]
KALA Polylysine – [57]
KALA (Poly(DMAEMA-NVP))-b-PEG-galactose Galactose [56]
46, Hel 11-7 – – [68]
INF7, JTS-1 K8 peptide K(GalNAc)2 [69]
PpTG1, ppTG20 – – [64]
LAH4 analogues – – [62]
a
Neutral lipid DOPE was added into the complex for transfection enhancement.

nents and GALA’s ability to destabilize membranes Gramicidin S strongly binds to DNA by charge
at low pH. interactions and mediates high level of luciferase
Gene transfection of a formulation based on DNA/ transfection activity ( f 109 light units per mg of cell
cationic liposome/transferrin complex can be en- protein) [11]. More interestingly, the peptide/DNA/
hanced by GALA peptide [52 – 54]. By studying the DOPE complex mediates rapid association of DNA
transfection mechanism of the complex, the presence with cells, and lysomotrophic agents do not influence
of transferrin is proposed to trigger internalization of the extent of transfection, which suggest that DNA
the complex by receptor-mediated endocytosis and the entry into the cell may be directly via the plasma
GALA peptide is used to promote endosomal desta- membrane [11]. Our unpublished in vivo studies with
bilization and release the genetic material into cyto- this complex indicated a substantial level of toxicity
plasm. Since the size of the pores (5– 10 Å) caused by when the gramicidin S/DOPE/DNA complexes were
GALA is too small to allow the direct passage of injected intramuscularly into animals.
DNA through, GALA was suggested to not only KALA is a fusogenic peptide designed to fulfill all
induce the membrane disruption at low pH but also the aspects required for efficient gene delivery without
promote the DNA unpacking [54]. These studies the aid of other components [12]. To our knowledge,
support the concept that pH-sensitive membrane dis- KALA was the first designed peptide, which can bind
rupting peptides such as GALA can improve gene DNA, destabilize membranes and mediate significant
transfer. To date, the interaction mechanism between gene delivery. KALA undergoes a pH-dependent
GALA and the DNA condensing reagent (including amphipathic a-helix to random coil conformational
polylysine, cationic liposome or dendrimer) is not change when environmental pH decreased from 7.5 to
well characterized and requires further evaluation if 5.0. KALA mediates release of entrapped dyes from
fusogenic peptide-based gene delivery systems are to lipid vesicles in the presence and absence of ODN,
be improved (Table 1). and the leakage is correlated with interaction of the
Compared to the multi-component systems de- peptide with bilayer by KALA tryptophan emission
scribed above, the cyclic amphipathic peptide (gram- spectrum [12]. Both ODN and plasmid DNA can be
icidin S) a cyclic h-turn structure was used to form a delivered into cells by KALA. GALA, by itself, on the
three-component gene delivery system [11]. In this other hand did not enhance the delivery due to its
system, the amphipathic peptide itself is a DNA- strong negative charge. This demonstrated that the
condensing reagent; the only other component in- positively charged Lys residues on KALA are re-
volved in, is the fusogenic ‘‘helper’’ lipid, DOPE. quired for DNA (or ODN) condensing and intracel-
982 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

lular trafficking. However, the mechanism of how more hydrophilic character, such as the polyhistidine
KALA promotes the transfer of DNA across mem- peptides [59] may be able to buffer the pH in endo-
branes is unknown. somes and promote a lysosomotropic effect [18,60]
KALA has also be used with other components, but are unable to disrupt membranes unless hydro-
such as polyethylenimine (PEI) [55], (poly phobic residues are included [13,61,62].
(DMAEMA-NVP))-b-PEG-galactose [56], polylysine Whether or not the a-helical motif is the preferred
[57] to formulate DNA complex for gene delivery secondary structure for membrane disruption, is not
(Table 1). In the KALA/polylysine/DNA system, known. Other structural motifs such as the hydropho-
KALA increases the transfection level of luciferase bic a-helix or the h-sheet have not been well studied
activity 10,000 times in evaluated conditions, and because of the difficulties to make them water soluble
further displacement of native polylysine to poly(eth- in the case of the hydrophobic a-helix and the prob-
ylene glycol) (PEG) modified polylysine keeps the lems associated with the propensity of hydrophobic h-
delivery system at high transfection level but obvi- sheet structures to form insoluble fibers.
ously decreases the toxicity [57]. PEGylated KALA is A drawback of the peptide membrane destabilizers
used to coat the surface of DNA/PEI complex to is that current sequences range in length from 9 to 30
prevent the nonspecific aggregation of particles and amino acids. This is not a problem for an experimental
enhance the gene delivery efficacy by membrane delivery system but if such molecules are to have a
destabilization [55]. commercial role, the therapeutic agent being delivered
must either be very potent or produce a therapeutic
effect that cannot be achieved using other technolo-
5. What are the advantages of GALA-like peptides gies. This is because if the peptides are a significant
as components of a drug and gene delivery system? fraction of the dose, the cost of the components must
be factored into the final cost of the product. Thus less
We believe that the advantages of synthetic pep- complicated synthetic molecules might be devised to
tides as components of delivery systems are that they: perform functions analogous to the peptides.
can be synthesized in high purity, are biocompatible A variety of groups have synthesized a series of
and have a significant data base for the secondary peptides to optimize gene delivery [62 – 64]. The
structural preference and hydrophobic contribution of highly refined tools of combinational chemistry and
the side chains. Moreover, there is an increasing high throughput screening system make it possible to
availability of interesting side chains that can be select suitable peptide-based gene delivery system
incorporated into a peptide to endow the peptide with from a large library of peptide analogues in the
useful characteristics. Finally, combinatorial synthetic absence of defined sequence– structure –function re-
methods and high throughput screening enable the lationship [65]. The challenge for the field is to design
identification of interesting sequences that improve screens that will predict how such systems work for in
cytoplasmic delivery. vivo gene transfer.
Clearly the low pH-sensitive amphipathic a-helical
structural motif lends itself to incorporation into water
soluble complexes or onto the surface of liposomes 6. Conclusions
precisely because such peptides can be manipulated in
water at neutral pH. The advantage of a titratable Studies with the pH-sensitive peptide GALA and
linkage compared to a hydrolytically or an enzymat- its analogues have been particularly informative, es-
ically activated linkage is that the rate of conversion is pecially regarding the requirement of a long amphi-
orders of magnitude faster for the titration compared pathic a-helical conformation to efficiently perturb
to the other two mechanisms [58]. These biorespon- lipid membranes. This phenomenon raised the possi-
sive peptides exploit the pH-drop as the endosome bility to include the GALA peptide for gene delivery
acidifies to between pH 5 and 6. Peptides with a more where the pH-sensitive peptide can perturb the cell
hydrophobic character, such as the influenza based membrane system to help the vector-encapsulated
fusion peptides have solubility limits. Peptides with a nucleic acid drugs escaping from endosome in low
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 983

pH environment. Since GALA has net negative [3] N.K. Subbarao, C.J. Fielding, R.L. Hamilton, F.C. Szoka Jr.,
charges, it is usually used together with other DNA Lecithin:cholesterol acyltransferase activation by synthetic
amphipathic peptides, Proteins 3 (1988) 187 – 198.
or ODN condensing reagents to formulate particles for [4] R.A. Parente, L. Nadasdi, N.K. Subbarao, F.C. Szoka Jr., Asso-
delivery. Amphipathic peptides other than GALA also ciation of a pH-sensitive peptide with membrane vesicles: role
mediate high efficient gene transfection. A partial of amino acid sequence, Biochemistry 29 (1990) 8713 – 8719.
replacement of Glu by Lys in the GALA sequence [5] R.A. Parente, S. Nir, F.C. Szoka Jr., Mechanism of leakage of
phospholipid vesicle contents induced by the peptide GALA,
yields the KALA peptide with a pH-dependent ability
Biochemistry 29 (1990) 8720 – 8728.
to form pores drastically different from that of GALA. [6] E. Fattal, S. Nir, R.A. Parente, F.C. Szoka Jr., Pore-forming
The cationic peptide KALA also has the membrane- peptides induce rapid phospholipid flip-flop in membranes,
perturbing capabilities, and its additional ability is to Biochemistry 33 (1994) 6721 – 6731.
condense DNA, which can be used to deliver ODN [7] F. Nicol, S. Nir, F.C. Szoka Jr., Effect of cholesterol and
and plasmid DNA into the nucleus of cells without charge on pore formation in bilayer vesicles by a pH-sensitive
peptide, Biophys. J. 71 (1996) 3288 – 3301.
other condensing reagent although the conformation [8] F. Nicol, S. Nir, F.C. Szoka Jr., Orientation of the pore-forming
change of KALA is from a-helix to random coil when peptide GALA in POPC vesicles determined by a BODIPY-
the pH decreased from neutral to the acidic range. avidin/biotin binding assay, Biophys. J. 76 (1999) 2121 – 2141.
The mechanisms of membrane perturbation in- [9] F. Nicol, S. Nir, F.C. Szoka Jr., Effect of phospholipid com-
duced by amphipathic a-helical peptide-based gene position on an amphipathic peptide-mediated pore formation
in bilayer vesicles, Biophys. J. 78 (2000) 818 – 829.
delivery systems are not yet understood and explan- [10] S. Nir, F. Nicol, F.C. Szoka Jr., Surface aggregation and mem-
ations for how such systems function to deliver DNA brane penetration by peptides: relation to pore formation and
into cells is based upon work in synthetic lipid fusion, Mol. Membr. Biol. 16 (1999) 95 – 101.
vesicles. This is not a criticism just of the peptide [11] J.Y. Legendre, F.C. Szoka Jr., Cyclic amphipathic peptide-
DNA complexes mediate high-efficiency transfection of ad-
systems rather it is a shortcoming of almost all of the
herent mammalian cells, Proc. Natl. Acad. Sci. U. S. A. 90
non-viral gene delivery systems. Further work is (1993) 893 – 897.
necessary to gain insights into the mechanisms of [12] T.B. Wyman, F. Nicol, O. Zelphati, P.V. Scaria, C. Plank, F.C.
membrane perturbation by amphipathic a-helices Szoka Jr., Design, synthesis, and characterization of a cationic
when they are complexed with DNA or ODN, in peptide that binds to nucleic acids and permeabilizes bilayers,
living cells to insure that studies in lipid vesicles are a Biochemistry 36 (1997) 3008 – 3017.
[13] C. Plank, M.X. Tang, A.R. Wolfe, F.C. Szoka Jr., Branched
good indicator for what is occurring when a delivery cationic peptides for gene delivery: role of type and number of
system interacts with cells. cationic residues in formation and in vitro activity of DNA
polyplexes, Hum. Gene Ther. 10 (1999) 319 – 332.
[14] M.C. Morris, L. Chaloin, F. Heitz, G. Divita, Translocating
peptides and proteins and their use for gene delivery, Curr.
Acknowledgements Opin. Biotechnol. 11 (2000) 461 – 466.
[15] K. Matsuzaki, Magainins as paradigm for the mode of action
Research on GALA in the Szoka laboratory has of pore forming polypeptides, Biochim. Biophys. Acta 1376
been supported by the National Institutes of Health, (1998) 391 – 400.
[16] J. Gariepy, K. Kawamura, Vectorial delivery of macromole-
most recently by R01 EB003008. Mass spectrometry
cules into cells using peptide-based vehicles, Trends Biotech-
analysis of peptide sequences was supported by nol. 19 (2001) 21 – 28.
NCRR RR01614 (UCSF). [17] R.A. Stull, G. Zon, F.C. Szoka Jr., Single-stranded phospho-
diester and phosphorothioate oligonucleotides bind actinomy-
cin D and interfere with tumor necrosis factor-induced lysis in
the L929 cytotoxicity assay, Antisense Res. Dev. 3 (1993)
References 295 – 300.
[18] J. Haensler, F.C. Szoka Jr., Polyamidoamine cascade polymers
[1] N.K. Subbarao Jr., R.A. Parente, F.C. Szoka Jr., L. Nadasdi, mediate efficient transfection of cells in culture, Bioconjug.
K. Pongracz, pH-dependent bilayer destabilization by an am- Chem. 4 (1993) 372 – 379.
phipathic peptide, Biochemistry 26 (1987) 2964 – 2972. [19] I. Auger, Computational techniques to predict amphipathic
[2] R.A. Parente, S. Nir, F.C. Szoka Jr., pH-dependent fusion of helical segments, in: R. Epand (Ed.), The Amphipathic Helix,
phosphatidylcholine small vesicles. Induction by a synthetic CRC Press, Boca Raton, 1993, pp. 8 – 19.
amphipathic peptide, J. Biol. Chem. 263 (1988) 4724 – 4730. [20] P.C. Biggin, M.S. Sansom, Interactions of alpha-helices with
984 W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985

lipid bilayers: a review of simulation studies, Biophys. Chem. [40] L. Bungener, A. Huckriede, J. Wilschut, T. Daemen, Delivery
76 (1999) 161 – 183. of protein antigens to the immune system by fusion-active
[21] Y. Shai, Mechanism of the binding, insertion and destabiliza- virosomes: a comparison with liposomes and ISCOMs, Bio-
tion of phospholipid bilayer membranes by alpha-helical anti- sci. Rep. 22 (2002) 323 – 338.
microbial and cell non-selective membrane-lytic peptides, [41] J.D. Lear, Z.R. Wasserman, W.F. DeGrado, Synthetic amphi-
Biochim. Biophys. Acta 1462 (1999) 55 – 70. philic peptide models for protein ion channels, Science 240
[22] R.B. Cornell, How cytidylyltransferase uses an amphipathic (1988) 1177 – 1181.
helix to sense membrane phospholipid composition, Biochem. [42] A. Kim, F.C. Szoka Jr., Amino acid side-chain contributions to
Soc. Trans. 26 (1998) 539 – 544. free energy of transfer of tripeptides from water to octanol,
[23] B. Bechinger, Understanding peptide interactions with the lipid Pharm. Res. 9 (1992) 504 – 514.
bilayer: a guide to membrane protein engineering, Curr. Opin. [43] E. Goormaghtigh, J. De Meutter, F. Szoka, V. Cabiaux, R.A.
Chem. Biol. 4 (2000) 639 – 644. Parente, J.M. Ruysschaert, Secondary structure and orienta-
[24] G. Menestrina, Escherichia coli hemolysin permeabilizes tion of the amphipathic peptide GALA in lipid structures. An
small unilamellar vesicles loaded with calcein by a single- infrared-spectroscopic approach, Eur. J. Biochem. 195 (1991)
hit mechanism, FEBS Lett. 232 (1988) 217 – 220. 421 – 429.
[25] M. Zasloff, Antimicrobial peptides of multicellular organisms, [44] C. Puyal, L. Maurin, G. Miquel, A. Bienvenue, J. Philippot,
Nature 415 (2002) 389 – 395. Design of a short membrane-destabilizing peptide covalently
[26] M.S. Sansom, The biophysics of peptide models of ion chan- bound to liposomes, Biochim. Biophys. Acta 1195 (1994)
nels, Prog. Biophys. Mol. Biol. 55 (1991) 139 – 235. 259 – 266.
[27] M.S. Sansom, Alamethicin and related peptaibols—model ion [45] I.A. Wilson, J.J. Skehel, D.C. Wiley, Structure of the haemag-
channels, Eur. Biophys. J. 22 (1993) 105 – 124. glutinin membrane glycoprotein of influenza virus at 3 Å
[28] K. He, S.J. Ludtke, W.T. Heller, H.W. Huang, Mechanism of resolution, Nature 289 (1981) 366 – 373.
alamethicin insertion into lipid bilayers, Biophys. J. 71 (1996) [46] P.A. Bullough, F.M. Hughson, J.J. Skehel, D.C. Wiley, Struc-
2669 – 2679. ture of influenza haemagglutinin at the pH of membrane fu-
[29] M.S. Sansom, Ion channels: structure of a molecular brake, sion, Nature 371 (1994) 37 – 43.
Curr. Biol. 9 (1999) R173 – R175. [47] N. Emans, J. Biwersi, A.S. Verkman, Imaging of endosome
[30] G.R. Smith, M.S. Sansom, Free energy of a potassium ion in a fusion in BHK fibroblasts based on a novel fluorimetric avi-
model of the channel formed by an amphipathic leucine – ser- din – biotin binding assay, Biophys. J. 69 (1995) 716 – 728.
ine peptide, Eur. Biophys. J. 31 (2002) 198 – 206. [48] C. Plank, K. Zatloukal, M. Cotten, K. Mechtler, E. Wagner,
[31] S. Oiki, V. Madison, M. Montal, Bundles of amphipathic Gene transfer into hepatocytes using asialoglycoprotein recep-
transmembrane alpha-helices as a structural motif for ion-con- tor mediated endocytosis of DNA complexed with an artificial
ducting channel proteins: studies on sodium channels and tetra-antennary galactose ligand, Bioconjug. Chem. 3 (1992)
acetylcholine receptors, Proteins 8 (1990) 226 – 236. 533 – 539.
[32] R.M. Stroud, M.P. McCarthy, M. Shuster, Nicotinic acetylcho- [49] K. Zatloukal, E. Wagner, M. Cotten, S. Phillips, C. Plank, P.
line receptor superfamily of ligand-gated ion channels, Bio- Steinlein, D.T. Curiel, M.L. Birnstiel, Transferrinfection: a
chemistry 29 (1990) 11009 – 11023. highly efficient way to express gene constructs in eukaryotic
[33] R. Henderson, J.M. Baldwin, T.A. Ceska, F. Zemlin, E. cells, Ann. New York Acad. Sci. 660 (1992) 136 – 153.
Beckmann, K.H. Downing, Model for the structure of bacter- [50] E. Wagner, C. Plank, K. Zatloukal, M. Cotten, M.L. Birnstiel,
iorhodopsin based on high-resolution electron cryo-micro- Influenza virus hemagglutinin HA-2 N-terminal fusogenic pep-
scopy, J. Mol. Biol. 213 (1990) 899 – 929. tides augment gene transfer by transferrin – polylysine – DNA
[34] M.D. Marger, M.H. Saier Jr., A major superfamily of trans- complexes: toward a synthetic virus-like gene-transfer vehicle,
membrane facilitators that catalyse uniport, symport and anti- Proc. Natl. Acad. Sci. U. S. A. 89 (1992) 7934 – 7938.
port, Trends Biochem. Sci. 18 (1993) 13 – 20. [51] C. Plank, B. Oberhauser, K. Mechtler, C. Koch, E. Wagner,
[35] J.J. Wine, Cystic fibrosis. Indictment of pore behaviour, Na- The influence of endosome-disruptive peptides on gene trans-
ture 366 (1993) 18 – 19. fer using synthetic virus-like gene transfer systems, J. Biol.
[36] B. Bechinger, Structure and functions of channel-forming pep- Chem. 269 (1994) 12918 – 12924.
tides: magainins, cecropins, melittin and alamethicin, J. Membr. [52] S. Simoes, V. Slepushkin, R. Gaspar, M.C. de Lima, N. Duz-
Biol. 156 (1997) 197 – 211. gunes, Gene delivery by negatively charged ternary complexes
[37] T. Stegmann, Membrane fusion mechanisms: the influenza of DNA, cationic liposomes and transferrin or fusigenic pep-
hemagglutinin paradigm and its implications for intracellular tides, Gene Ther. 5 (1998) 955 – 964.
fusion, Traffic 1 (2000) 598 – 604. [53] S. Simoes, V. Slepushkin, E. Pretzer, P. Dazin, R. Gaspar,
[38] J.M. White, Viral and cellular membrane fusion proteins, An- M.C. Pedroso de Lima, N. Duzgunes, Transfection of human
nu. Rev. Physiol. 52 (1990) 675 – 697. macrophages by lipoplexes via the combined use of transfer-
[39] P.M. Fischer, E. Krausz, D.P. Lane, Cellular delivery of im- rin and pH-sensitive peptides, J. Leukocyte Biol. 65 (1999)
permeable effector molecules in the form of conjugates with 270 – 279.
peptides capable of mediating membrane translocation, Bio- [54] S. Simoes, V. Slepushkin, P. Pires, R. Gaspar, M.P. de Lima,
conjug. Chem. 12 (2001) 825 – 841. N. Duzgunes, Mechanisms of gene transfer mediated by lip-
W. Li et al. / Advanced Drug Delivery Reviews 56 (2004) 967–985 985

oplexes associated with targeting ligands or pH-sensitive pep- [63] D. Balicki, C.D. Putnam, P.V. Scaria, E. Beutler, Structure
tides, Gene Ther. 6 (1999) 1798 – 1807. and function correlation in histone H2A peptide-mediated
[55] H. Lee, J.H. Jeong, T.G. Park, A new gene delivery formula- gene transfer, Proc. Natl. Acad. Sci. U. S. A. 99 (2002)
tion of polyethylenimine/DNA complexes coated with PEG 7467 – 7471.
conjugated fusogenic peptide, J. Control. Release 76 (2001) [64] K. Rittner, A. Benavente, A. Bompard-Sorlet, F. Heitz, G.
183 – 192. Divita, R. Brasseur, E. Jacobs, New basic membrane-destabi-
[56] D.W. Lim, Y.I. Yeom, T.G. Park, Poly(DMAEMA-NVP)-b- lizing peptides for plasmid-based gene delivery in vitro and in
PEG-galactose as gene delivery vector for hepatocytes, Bio- vivo, Mol. Ther. 5 (2002) 104 – 114.
conjug. Chem. 11 (2000) 688 – 695. [65] J.E. Murphy, T. Uno, J.D. Hamer, F.E. Cohen, V. Dwarki,
[57] H. Lee, J.H. Jeong, T.G. Park, PEG grafted polylysine with R.N. Zuckermann, A combinatorial approach to the discovery
fusogenic peptide for gene delivery: high transfection effi- of efficient cationic peptide reagents for gene delivery, Proc.
ciency with low cytotoxicity, J. Control. Release 79 (2002) Natl. Acad. Sci. U. S. A. 95 (1998) 1517 – 1522.
283 – 291. [66] M.J. Turk, J.A. Reddy, J.A. Chmielewski, P.S. Low, Charac-
[58] X. Guo, F.C. Szoka, Chemical approaches to triggerable lipid terization of a novel pH-sensitive peptide that enhances drug
vesicles for drug and gene delivery, Acc. Chem. Res. 36 (2003) release from folate-targeted liposomes at endosomal pHs, Bio-
335 – 341. chim. Biophys. Acta 1559 (2002) 56 – 68.
[59] D.W. Pack, D. Putnam, R. Langer, Design of imidazole-con- [67] J.H. Jeong, S.W. Kim, T.G. Park, Novel intracellular delivery
taining endosomolytic biopolymers for gene delivery, Biotech- system of antisense oligonucleotide by self-assembled hybrid
nol. Bioeng. 67 (2000) 217 – 223. micelles composed of DNA/PEG conjugate and cationic fuso-
[60] O. Boussif, F. Lezoualc’h, M.A. Zanta, M.D. Mergny, D. genic peptide, Bioconjug. Chem. 14 (2003) 473 – 479.
Scherman, B. Demeneix, J.P. Behr, A versatile vector for gene [68] T. Niidome, K. Takaji, M. Urakawa, N. Ohmori, A. Wada, T.
and oligonucleotide transfer into cells in culture and in vivo: Hirayama, H. Aoyagi, Chain length of cationic alpha-helical
polyethylenimine, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) peptide sufficient for gene delivery into cells, Bioconjug.
7297 – 7301. Chem. 10 (1999) 773 – 780.
[61] C. Pichon, C. Goncalves, P. Midoux, Histidine-rich peptides [69] S.M. Van Rossenberg, K.M. Sliedregt-Bol, N.J. Meeuwenoord,
and polymers for nucleic acids delivery, Adv. Drug Deliv. T.J. Van Berkel, J.H. Van Boom, G.A. Van Der Marel, E.A.
Rev. 53 (2001) 75 – 94. Biessen, Targeted lysosome disruptive elements for improve-
[62] A. Kichler, C. Leborgne, J. Marz, O. Danos, B. Bechinger, ment of parenchymal liver cell-specific gene delivery, J. Biol.
Histidine-rich amphipathic peptide antibiotics promote effi- Chem. 277 (2002) 45803 – 45810.
cient delivery of DNA into mammalian cells, Proc. Natl.
Acad. Sci. U. S. A. 100 (2003) 1564 – 1568.

You might also like