Sie sind auf Seite 1von 14

Blackwell Science, LtdOxford, UKCMICellular Microbiology 1462-5814Blackwell Publishing Ltd, 200571014181431Original ArticleEntry domain of azurinT. Yamada et al.

Cellular Microbiology (2005) 7(10), 14181431 doi:10.1111/j.1462-5822.2005.00567.x

Internalization of bacterial redox protein azurin in


mammalian cells: entry domain and specificity

Tohru Yamada,1,2 Arsenio M. Fialho,1 Vasu Punj,2 often dictated by a small segment of the protein, which
Laura Bratescu,2 Tapas K. Das Gupta2 and can then be used as a vehicle to transport foreign proteins
Ananda M. Chakrabarty1* into such cells. Amphipathic peptides are used to facilitate
Departments of 1Microbiology and Immunology and uptake of DNA-cleaving metalloporphyrins as potential
2
Surgical Oncology, University of Illinois College of antitumour drugs in human fibroblasts HS68 or murine
Medicine, Chicago, IL 60612, USA. lymphocytic leukaemia L1210 cells (Chaloin et al., 2001).
Peptides, called cell-penetrating peptides, such as pene-
tratin, transportan, Tat (amino acids 4757 or 4860) and
Summary
the model amphipathic peptide MAP, have been widely
Azurin is a member of a group of copper-containing used as delivery vehicles for transporting pharmacologi-
redox proteins called cupredoxins. Different cupre- cally important substances such as antisense oligonu-
doxins are produced by different aerobic bacteria as clotides, proteins and peptides (Lindgren et al., 2000;
agents of electron transfer. Recently, we demon- Hallbrink et al., 2001). Such peptides, particularly the
strated that azurin enters into J774 and several types DNA-binding homeodomain of Antennapedia, a Droso-
of cancer cells leading to the induction of apoptosis. phila transcription factor, or the 21-residue peptide carrier
We now demonstrate that azurin is internalized in Pep-1, are internalized by many types of cells in culture
J774 or cancer cells in a temperature-dependent man- such as human HS68 or murine NIH-3T3 fibroblasts either
ner. Azurin shows preferential entry into cancer com- at 37C or at 4C, suggesting a penetration mechanism
pared with normal cells. An 28-amino-acid fragment different from the classical endocytosis (Morris et al.,
of azurin fused to glutathione S-transferase (GST) or 2001) and which requires no chiral receptor proteins. One
the green fluorescent protein (GFP), which are inca- of the most widely used peptides to transport pharmaco-
pable of entering mammalian cells by themselves, can logically active compounds in mammalian cells is the 11-
be internalized in J774 or human melanoma or breast amino-acid arginine-rich sequence (PTD, protein trans-
cancer cells at 37C, but not at 4C. Competition duction domain) of the human immunodeficiency virus
experiments as well as studies with inhibitors such type 1 (HIV-1) transactivator protein Tat (Schwarze et al.,
as cytochalasin D suggest that azurin may enter cells, 1999; 2000). Intraperitoneal injection of the 120 kDa b-
at least in part, by a receptor-mediated endocytic pro- galactosidase/Tat fusion protein results in the transcellular
cess. The 28-amino-acid peptide therefore acts as a transduction of the fusion protein into virtually all tissues
potential protein transduction domain (PTD), and can in mice, including the passage of the bloodbrain barrier
be used as a vehicle to transport cargo proteins such (Schwarze et al., 1999). This short peptide domain of HIV-
as GST and GSTGFP fusion proteins. Another mem- 1 Tat has been shown to mediate cell internalization of
ber of the cupredoxin family, rusticyanin, that has also large molecules or particles, including magnetic nanopar-
been shown to enter J774 and human cancer cells and ticles, phage vectors, liposomes and plasmid DNA.
exert cytotoxicity, does not demonstrate preferential Interestingly, unlike the other cell-penetrating peptides,
entry for cancer cells and lacks the structural features internalization of cargo proteins by full-length Tat or its 11-
characteristic of the azurin PTD. amino-acid transduction domain is significantly impaired
at 4C (Liu et al., 2000; Suzuki et al., 2002) and requires
interactions with receptors such as the heparan sulphate
Introduction
chains of the cell membrane heparan sulphate proteogly-
Much attention has recently been directed towards an cans (Tyagi et al., 2001). The use of the PTD of the Tat
understanding of how proteins or peptides enter mamma- protein in fusion with a peptide derived from the C-terminal
lian cells. The entry of a protein into a mammalian cell is domain of the tumour suppressor protein p53 (p53C
peptide) in its D-isomeric and inverted sequence (RI-
TATp53C) has been shown to allow activation of p53 in
Received 1 April, 2005; revised 29 April, 2005; accepted 3 May, 2005.
*For correspondence. E-mail pseudomo@uic.edu; Tel. (+1) cancer but not in normal cells. Treatment by RI-TATp53C
312 996 4586; Fax (+1) 312 996 6415. peptide of pre-clinical terminal peritoneal carcinomatosis

2005 Blackwell Publishing Ltd


Entry domain of azurin 1419
and peritoneal lymphoma models results in significant charged amino acids, had reduced ability to induce apo-
increase in life span and generation of disease-free ani- ptosis in J774 cells but had an enhanced ability to allow
mals (Snyder et al., 2004), thus demonstrating potential inhibition of cell cycle progression at the G1 to S phase
interesting applications of the PTDs in cancer therapy. The in J774 cells (Yamada et al., 2004a). However, very little
internalization of cargo proteins fused to the Tat PTD is is known about the mode of entry of bacterial redox pro-
impaired at 4C, suggesting the role of an active endocytic teins such as azurin and cytochrome c551 in mammalian
process (Tyagi et al., 2001) and indeed such a process cells. We report the domain of azurin that is involved in its
has been shown to originate from the cell membrane lipid entry to J774 or UISO-Mel-2 cells and some of the char-
rafts involving caveolar endocytosis (Fittipaldi et al., 2003). acteristics of the entry domain, including its ability to trans-
Most of the PTDs to date have been derived from viral port foreign proteins such as glutathione S-transferase
and mammalian sources (Hallbrink et al., 2001; Morris (GST) or enhanced green fluorescent protein (GFP)
et al., 2001). Although bacterial proteins such as cholera inside such cells.
toxin are known to enter mammalian cell cytosol (Sofer
and Futerman, 1995), the cytotoxicity of such proteins has
limited the use of bacterial proteins, or PTDs derived from Results
them, for transporting pharmacologically important cargos
Entry of cupredoxins in mammalian cells
in mammalian cells. We have recently described the
secretion of two redox proteins, azurin and cytochrome We previously demonstrated that azurin entered both
c551, by Pseudomonas aeruginosa that enter mammalian J774 (Yamada et al., 2002a) and human cancer (Yamada
cells such as J774 cell line-derived macrophages and et al., 2002b; Punj et al., 2004) cells. However, it was not
induce apoptotic cell death (Zaborina et al., 2000). On clear whether azurin was unique in this respect or whether
entry into J774 cells, azurin localizes in the cytosol and other purified cupredoxins might also enter mammalian
nuclear fractions, and forms a complex with the tumour cells. The secretion of azurin by P. aeruginosa, a known
suppressor protein p53, thereby stabilizing it and enhanc- opportunistic pathogen, also implied that azurin might act
ing its intracellular level (Yamada et al., 2002a). A higher as a virulence factor because of its ability to induce apo-
intracellular level of p53 then triggers apoptosis. ptotic death in macrophage-like J774 cells (Zaborina
Redox-negative apo-azurin devoid of copper or some et al., 2000). J774 cells are, however, not normal primary
site-directed mutants of azurin defective in electron macrophages but are ascites forms of murine reticulum
transfer could form complexes with p53 and were found cell sarcoma with macrophage-like properties of adher-
to be cytotoxic. A redox-negative site-directed mutant ence, morphology, receptors for immunoglobulin and anti-
(M44KM64E), however, appeared to interfere in the oligo- body-dependent lysis of target cells (Ralph and Nakoinz,
merization of p53 and demonstrated very little cytotoxicity. 1975). We previously reported that while azurin induced
This suggested that the redox activity of azurin was not apoptosis in vivo in the tumour cells in nude mice, it
important for its cytotoxicity (Goto et al., 2003). The induc- showed much less cytotoxicity towards normal tissues in
tion of azurin-mediated apoptosis was not limited to J774 azurin-treated mice (Yamada et al., 2002b; Punj et al.,
cells. Azurin could also enter cancer cells such as human 2004) and indeed in in vitro studies, azurin showed much
melanoma UISO-Mel-2 (Yamada et al., 2002b) or human less cytotoxicity in normal human mammary epithelial
breast cancer MCF-7 (Punj et al., 2004) cells. In both cells such as MCF-10F than in MCF-7 breast cancer cells
cases, azurin allowed the elevation of the intracellular p53 (Punj et al., 2004). It was therefore of interest to examine
levels, leading to enhanced Bax formation and induction whether other cupredoxins besides azurin, particularly
of apoptosis in such cells. Most interestingly, intraperito- those from non-pathogenic bacteria such as the acido-
neal injection of azurin in nude mice harbouring philic chemolithotroph Acidithiobacillus ferrooxidans
xenografted Mel-2 or MCF-7 human cancers led to statis- which grows well at pH values 2.03.5 and produces
tically significant regression of such cancers (Yamada rusticyanin as an electron transfer agent during oxidation
et al., 2002b; Punj et al., 2004), suggesting the potential of ferrous iron (Fe2+) to ferric iron (Fe3+) (Appia-Ayme
use of azurin in cancer therapy. Cytochrome c551, on the et al., 1999; Grossman et al., 2002), will enter J774 and
other hand, could enter J774 cells but induced inhibition other mammalian cells to demonstrate cytotoxicity. The
of cell cycle progression at the G1 to S phase (Hiraoka entry of wild-type (WT) azurin and another cupredoxin
et al., 2004). A mutant form of cytochrome c551, V23DI59E rusticyanin, which has also been shown to possess cyto-
cytochrome c551, while having reduced ability to trigger toxicity (Yamada et al., 2004b), in J774 cells is shown in
inhibition of cell cycle progression, allowed induction of Fig. 1A. In these experiments, the cupredoxins were con-
apoptosis in J774 cells (Hiraoka et al., 2004). Interest- jugated with Alexa fluor 568 to fluoresce red and incu-
ingly, a similar mutant form of azurin, M44KM64E azurin, bated with J774 cells for 1 h at 37C at a concentration of
where two hydrophobic amino acids were replaced by two 7 mM. The nucleus was stained blue with 4,6-diamidino-
2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431
1420 T. Yamada et al.

A B Fig. 1. Entry of bacterial cupredoxins in mam-


malian cells.
Control Azurin Rusticyanin 45 min 6h
A. Internalization of azurin and rusticyanin was
visualized by confocal microscopy. Both azurin
J774 and rusticyanin (7 mM) were labelled with red
fluorescent dye Alexa fluor 568 and incubated
with J774 cells for 1 h at 37C. The nucleus is
labelled blue with DAPI. The scale bar corre-
sponds to 20 micrometers.
Peritoneal B and C. Wild-type azurin conjugated with
C 45 min 6h
macrophages Alexa fluor 568 was incubated for 45 min or 6 h
with various mammalian cells including J774,
peritoneal macrophages, mast cells, human
MCF-7 breast cancer MCF-7 and human normal epi-
thelial MCF-10F cells as indicated. The inter-
Mast cells
nalization was visualized by confocal
microscopy.
D. Alexa fluor-conjugated azurin at 7 mM was
MCF-10F incubated with equal numbers of cells of vari-
ous cell lines such as Mel-2 and fibroblasts on

D E a coverslip at 37C for 0.5 and 3.5 h after which


Mel-2 Fibroblast Control 30 min 5h images were taken.
E. Induction of apoptosis in normal fibroblast
and MCF-10F cells after microinjection of
0.5 h Fibroblast
azurin in such cells. Azurin mixed with dextran
and Texas red was microinjected as described
previously (Punj et al., 2004). Fluorescent
Mel-2 Fibroblast images of the microinjected cells were taken
MCF-10F
with a Zeiss LSM 510 confocal laser micro-
3.5 h scope after 30 min and 5 h.

2-phenylindole (DAPI). A control without the proteins was nuclear DNA (labelled blue with DAPI) condensation and
maintained. In both cases, the cupredoxins were seen to fragmentation were observed in microinjected single cells
enter J774 cells in the cytosol (Fig. 1A). in 5 h but not during 30 min (Fig. 1E) incubation with
As J774 cells are ascitic forms of murine reticulum cell azurin, demonstrating azurins ability to induce apoptosis
sarcoma, it was of interest to determine whether a cupre- once inside normal cells.
doxin such as azurin can enter normal peritoneal mac-
rophages or mast cells. As previous studies (Punj et al.,
Defining the entry domain of azurin
2004) showed a reduced cytotoxic activity of azurin
towards normal breast MCF-10F cells as contrasted with To define azurin entry domain, we constructed a series of
the MCF-7 cells, it was also of interest to examine whether GST fusions of 128-amino-acid-long azurin truncated at
azurin could enter such cells equally well. While azurin the central region, the N- and the C-terminals (Fig. 2A),
could efficiently be internalized in J774 cells during 45 min and purified the fusion protein products (Fig. 2B). Using
incubation, it was internalized very inefficiently in perito- Alexa fluor 568-conjugated WT azurin, GST and GST
neal macrophages or mast cells (Fig. 1B). Even after 6 h azurin (GSTazu) fusion derivatives, we examined their
incubation, such cells showed only limited entry. Similarly, internalization in J774 cells at 37C during 1 h incubation.
while azurin efficiently entered the breast cancer cells The nucleus was stained blue with DAPI. While WT azurin
(MCF-7), it showed extremely reduced entry in the normal was internalized, GST remained at the periphery of the
mammary MCF-10F cells (Fig. 1C). A reduced level of cells and was not internalized (Fig. 3A). GSTazu 36128
azurin in normal cells might explain a low level of apopto- and GSTazu 3689 were internalized as was GSTazu
sis during in vitro treatment with MCF-10F cells (Punj 3677 (Fig. 3A). Further truncations, however, demon-
et al., 2004). Similar results were obtained with human strated that while GSTazu 5077 was internalized, GST
melanoma Mel-2 and normal fibroblast cells (Fig. 1D). To azu 3650 produced a punctate labelling, indicating a slow
demonstrate that indeed cellular entry is the major con- and inefficient entry and possible localization on the cell
straint in azurins ability to induce apoptosis in normal surface or in the endosomes. Further truncation of GST
cells, we microinjected azurin, as described previously azu 5077 to GSTazu 5066 and GSTazu 6777 dem-
(Punj et al., 2004), in fibroblast and MCF-10F cells onstrated punctate labelling, confirming that for efficient
(Fig. 1E) and looked for induction of apoptosis, leading to internalization, amino acids 5077 were important
nuclear DNA condensation and fragmentation. Significant (Fig. 3A).

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


Entry domain of azurin 1421

A
1 128
WT azurin
aecsvdiqgn dqmqfntnai tvdksckqft vnlshpgnlp knvmghnwvl staadmqgvv tdgmasgldk dylkpddsrv iahtkligsg ekdsvtfdvs klkegeqymf fctfpghsal mkgtltlk
14 kDa
36 128
1, GST-azu 36-128 GST--- pgnlp knvmghnwvl staadmqgvv tdgmasgldk dylkpddsrv iahtkligsg ekdsvtfdvs klkegeqymf fctfpghsal mkgtltlk
10 kDa+26 kDa
36 89
2, GST-azu 36-89 GST--- pgnlp knvmghnwvl staadmqgvv tdgmasgldk dylkpddsrv iahtkligs
5.7 kDa+26 kDa
36 77
3, GST-azu 36-77 GST--- pgnlp knvmghnwvl staadmqgvv tdgmasgldk dylkpdd
4.4 kDa+26 kDa
36 50
4, GST-azu 36-50 GST--- pgnlp knvmghnwvl
1.6 kDa+26 kDa B
50 77 (kDa) 1 2 3 4 5 6 7 8
5, GST-azu 50-77 185
GST--- l staadmqgvv tdgmasgldk dylkpdd
2.9 kDa+26 kDa
50 66
6, GST-azu 50-66
GST--- l staadmqgvv tdgmas 52
1.6 kDa+26 kDa
67 77
7, GST-azu 67-77
GST--- gldk dylkpdd 31
1.2 kDa+26 kDa
8, GST
26 kDa 17

Fig. 2. A. Schematic representation of various truncated azurin constructs marked 18 as derived from WT azurin. The detailed procedures for
the constructs are given under Experimental procedures. Various sizes of azu gene were fused at the 3 end of the gst gene in frame.
B. GSTazu fusion proteins were purified after cellular growth and lysis, loaded on SDS-PAGE and visualized by Coomassie blue staining.
GSTazu 3650 (lane 4), 27.6 kDa, shows slightly aberrant mobility compared with GSTazu 5077 (lane 5), 28.9 kDa.

Azurin internalization is an energy-requiring process (amino acids 5066) and a hydrophilic (amino acids 67
77) domain (Fig. 3B). While the amphipathic peptide could
To determine whether the internalization of azurin is medi-
transport GST inside J774 cells at 37C, the hydrophobic
ated by an energy-requiring active endocytic process, we
(amino acids 5066) or the hydrophilic (amino acids
also examined the internalization of WT azurin and the
6777) domains by themselves were not proficient in
GSTazu fusion derivatives in J774 cells incubated at
this regard even at 37C (Fig. 3A). Neither was active,
4C. The fact that the 28-amino-acid azu 5077 fused to
however, at 4C (Fig. 3B) so that GSTazu 5066 or
GST was internalized in J774 cells at 37C (Fig. 3A) sug-
GSTazu 6777 were not internalized at 4C, similar to
gested that this moiety could transport GST, which is not
the amphipathic GSTazu 5077 (Fig. 3B).
by itself internalized in J774 cells (Fig. 3A). At 4C, how-
ever, internalization of WT azurin inside J774 cells during
1 h incubation was significantly reduced. Similar impair-
Energy-dependent internalization of the GSTGFPazu
ment was also seen with GSTazu 36128 and GSTazu
5077 fusion protein in J774 and melanoma UISO-Mel-2
3689 (Fig. 3B). Interestingly, the shorter GSTazu
cells
3677 and GSTazu 5077 demonstrated more severe
impairment of internalization at 4C, suggesting that The ability of the azu 5077 moiety to allow internalization
extended sequences beyond amino acids 77 (amino acids of GST raised an interesting question: can this amphip-
7789 and beyond) may contribute to a slow internaliza- athic azurin peptide allow internalization of even higher-
tion process at 4C, presumably via a cell-penetrating molecular-weight proteins and will the process be energy-
mechanism that does not require receptor-mediated dependent? To reduce costs, as well as any effect of Alexa
endocytosis (Lindgren et al., 2000; Hallbrink et al., 2001). fluor 568 conjugated to the fusion proteins, we fused GST
There is now evidence that the C-terminal part of azurin with GFP to make a GSTGFP fusion derivative. Addition-
has structural similarities with some mammalian surface ally, we fused azu 5077 moiety to the GSTGFP (molec-
protein ligands that bind surface-exposed receptors that ular mass 53 kDa) fusion protein (Fig. 4A) and examined
are highly expressed in cancer cells (Gough and Chothia, the mobility of the purified GST, GSTGFP and GST
2004), and azurin may also be internalized by binding with GFPazu 5077 fusion derivatives on SDS-PAGE, and
such receptors. It is interesting to note that residues 50 detected by Coomassie blue staining (Fig. 4B) and West-
77 is an amphipathic peptide with both a hydrophobic ern blotting using anti-azurin antibody (Fig. 4C).

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


1422 T. Yamada et al.

A PBS WT azurin GST GST-azu 36-128 GST-azu 36-89

GST-azu 36-77 GST-azu 36-50 GST-azu 50-77 GST-azu 50-66 GST-azu 67-77

PBS WT azurin GST GST-azu 36-128 GST-azu 36-89


B

GST-azu 36-77 GST-azu 50-77 GST-azu 50-66 GST-azu 67-77

50 67 77
lstaadmqgvvtdgmasgldkdylkpdd
3.0

-3.0

Fig. 3. Confocal microscopic images of J774 cells treated with 200 mg ml-1 GSTazu derivatives as depicted in Fig. 2A. Equal numbers of J774
cells were incubated with various GSTazu fusion proteins labelled with Alexa fluor 568. Cells were placed at 37C (A) or 4C (B) during protein
treatment (1 h) before visualization by confocal microscopy. Hydropathy plot of azu 5077 was generated by Kyte and Doolittle method with
GENETYX software.

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


Entry domain of azurin 1423

A D E
PBS GSTGFP GSTGFPazu 5077 PBS GSTGFP GSTGFPazu 5077
NH2-- GST GFP azu 5077 --COOH

7
-77
77

0-7
50-

u5
5 0
zu
FP
B C
GS urin

-az
rin

zu
P-a

FP
TG

azu
37C 37C

Ta
az

-GF

TG
T
WT

WT
GS

GS
GS

GS
(kDa) (kDa)
185 185
98 98

52 52

31 31
19 19
17
4C 4C
6 9
3
6

Fig. 4. Construction of GSTGFPazu 5077 fusion protein.


AC. (A) The gfp gene was introduced at the 3 end of gst gene (for GSTGFP) and azu 5077 oligonucleotide fragment was then ligated at the
3 end of the gfp gene in frame to produce GSTGFPazu 5077 fusion protein. GST, GSTGFP and GSTGFPazu 5077 were purified as
single fusion proteins from the cell lysates. Purified proteins, including WT azurin, were run on SDS-PAGE and detected by Coomassie blue
staining (B) and also by Western blotting using anti-azurin antibody (C). In (C), both monomeric azurin (14 kDa) and small amounts of a dimeric
form are seen. It should be noted that anti-azurin antibody does not cross-react with GST.
D and E. Temperature-dependence of GSTGFP or GSTGFPazu 5077 internalization in either J774 (D) or UISO-Mel-2 cells (E) cells. The
cells were treated with 4 mM GSTGFP or GSTGFPazu 5077 and incubated at 37C for 1 h. Green fluorescence derived from GSTGFP or
GSTGFPazu 5077 was visualized by confocal microscopy.

In order to see whether there is any difference in the assessment of the cargo-carrying PTD entry, we used
binding profile of GSTGFP and GSTGFPazu 5077, flow cytometry to measure GFP fluorescence, varying
we incubated both J774 and UISO-Mel-2 cells with GST either the concentrations of the fusion protein or the time
GFP and GSTGFPazu 5077 at 37C and at 4C and of incubation. Significant azurin PTD entry was observed
the green fluorescence was localized using confocal at concentrations above 1.0 mM and increasing entry was
microscopy. In J774 cells, GSTGFP fusion protein bound observed with increasing concentrations of the fusion pro-
to the surface and was not internalized both at 37C and tein in a linear manner up to 8 mM (Fig. 5A). The flow
at 4C (Fig. 4D). In contrast, GSTGFPazu 5077 was cytometric data were plotted diagrammatically as shown
found to be internalized at 37C but not at 4C, suggesting in the right hand panel of Fig. 5A. When 0.45 mM or
that the azu 5077 peptide could allow internalization of 1.8 mM fusion protein was used for entry determination at
the 53 kDa GSTGFP in J774 cells in a temperature- different periods of incubation, maximal entry was
dependent manner (Fig. 4D). In UISO-Mel-2 cells, the observed in about 1.0 h (Fig. 5B). As the entry of the
GSTGFP fusion protein was retained on the surface both fusion protein was shown to be low at 4C, implying the
at 37C and at 4C (Fig. 4E). In contrast, similar to J774 entry process (but not necessarily surface binding) as
cells, GSTGFPazu 5077 fusion protein was seen to energy-requiring (Fig. 4D and E), we tested the effect of
be internalized at 37C but not at 4C (Fig. 4E). Thus, the protonophore carbonyl cyanide m-chlorophenylhydra-
the azu 5077 peptide promotes internalization of a zone (CCCP), a mitochondrial uncoupler of energy gen-
53 kDa cargo protein in J774 or UISO-Mel-2 cells by an eration, both by flow cytometry (Fig. 5C) and by confocal
energy-dependent process, presumably mediated by an microscopy (using Alexa fluor-conjugated azurin; Fig. 5D,
endocytic pathway. panel CCCP, 20 mM) on azurin PTD (or intact azurin)
entry. There was substantial (about 40%) inhibition of
azurin PTD or azurin entry by CCCP under such condi-
Kinetics of the azurin PTD (GSTGFPazu 5077) entry
tions. We also tested the effect of two other inhibitors,
in J774 cells and the effect of inhibitors
cytochalasin D, a known inhibitor of receptor-mediated
The ability of the azurin PTD (azu 5077) to carry a endocytosis that disrupts the cellular microfilament net-
53 kDa cargo such as GSTGFP inside J774 or Mel-2 work, and Brefeldin A, which is known to disrupt the Golgi
cells (Fig. 4D and E) allowed us to study the kinetics of apparatus and inhibit classical vesicle-mediated secretion
entry of this fusion protein in J774 cells and some param- (Lippincott Schwartz et al., 1990; Elliott and OHare, 1997)
eters that affect this process. To obtain quantitative on the entry of Alexa fluor-conjugated azurin by confocal

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


1424 T. Yamada et al.

A B
100
GST-GFP-azu 50-77 (0.45 mM) 70

Events
Control GST-GFP-azu 50-77
80 Control 0.5 h 60

Positive cells (%)


0.9 mM

Positive cells (%)


1h
1.8 mM 3h
Events

3.6 mM 50
60
7.2 mM 40
40 GST-GFP-azu 50-77 (1.8 mM) 30

Events
Control 20
20 0.5 h
1h 10
3h
100 101 102 103 104 0
0 2 4 6 8 10 0
Green fluorescence GST-GFP-azu 50-77 (mM) 100 101 102 103 104 0 1 2 3
Green fluorescence Time (h)

C D Control CCCP Brefeldin A Cytochalasin D


70
20 mM 18 mM 0.25 mM 0.5 mM
60
Positive cells (%)

50
Events

40
GST-GFP-azu 50-77 + CCCP
30
GST-GFP-azu 50-77
20
10
100 101 102 103 104
0
Green fluorescence
GST-GFP-azu 50-77 + + +
CCCP 10 20 mM

Fig. 5. A. Kinetic study for internalization of GSTGFPazu 5077 fusion protein. Green fluorescence was assayed in J774 cells treated with
various concentrations of GSTGFPazu 5077 at 37C for 1 h. Ten thousand cells were analysed by flow cytometry. The untreated cells (control)
in flow cytogram constituted an internal negative control. The percentages of GSTGFPazu 5077-positive cells are represented in the right
panel.
B. Time-dependence of internalization of GSTGFPazu 5077. J774 cells were incubated with 0.45 mM (upper left panel and open circle in right
panel) or 1.8 mM (lower left panel and closed circle in right panel) GSTGFPazu 5077 for indicated times at 37C and collected by flow cytometry.
C. J774 cells were pre-treated with 0, 10 or 20 mM CCCP for 1 h and incubated with 1.8 mM GSTGFPazu 5077 for an additional hour at 37C.
Entry of GSTGFPazu 5077 in the presence (10 mM, red; 20 mM, blue) or absence (green) of CCCP was analysed by FACS.
D. Inhibition of azurin internalization by CCCP and cytochalasin D but not by Brefeldin A. Alexa fluor 568-conjugated azurin (14 mM) was incubated
with UISO-Mel-2 at 37C for 1 h in the presence of PBS (control) or specified concentrations of CCCP, Brefeldin A and cytochalasin D. The extent
of azurin internalization in each case was followed by confocal microscopy.

microscopy. While Brefeldin A had no effect, increasing conjugated GSTazu 5077 (7 mM) in the presence of 7,
concentrations of cytochalasin D caused significant inhi- 14 and 56 mM of unlabelled azurin for 1 h at 37C before
bition of azurin internalization (Fig. 5D), suggesting that determining GSTazu 5077 entry. The results clearly
azurin internalization is mediated, at least in part, by a demonstrated that in comparison with labelled 7 mM GST
receptor-mediated endocytic process. The modest inhibi- azu 5077 alone (Fig. 6A, top), the presence of increasing
tion of azurin internalization by CCCP or cytochalasin D amounts of unlabelled azurin (7, 14 and 56 mM; Fig. 6A,
implies that part of azurin is bound on the surface, pre- middle) increasingly competed with the entry of labelled
sumably due to azurins structural similarity at the C- 7 mM GSTazu 5077, thus demonstrating a common
terminal end with a group of mammalian proteins that are receptor binding by unlabelled azurin when used in
ligands for binding with a group of surface-exposed recep- excess. In contrast, when similar concentrations (6, 12
tor tyrosine kinases that are known to be hyperexpressed and 48 mM) of unlabelled rusticyanin (Fig. 6A, bottom)
in cancer cells (Gough and Chothia, 2004). were used in the presence of labelled GSTazu 5077,
very little effect on the entry of GSTazu 5077 was seen.
These results appear to imply that azurin entry involves
Azurin and rusticyanin use different modes of entry:
binding with receptors that do not recognize rusticyanin.
competition experiments and structural differences
To determine whether a lack of competition of the entry
To demonstrate that azurin internalization involves recep- of the azurin PTD by rusticyanin may imply a different
tor binding, we did a competition experiment with unla- mode of entry of rusticyanin, we checked the specificity
belled azurin at 37C in the presence of 7 mM Alexa fluor- of azurin and rusticyanin entries in cancer and normal
conjugated GSTazu 5077. We incubated J774 cells cells. As shown earlier (Fig. 1BD), Alexa fluor-
without Alexa fluor-conjugated azurin, with Alexa fluor- conjugated azurin entered efficiently in UISO-Mel-2 and
conjugated GSTazu 5077 (7 mM) and with Alexa fluor- MCF-7 cancer cells but not in the normal mammary MCF-

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


Entry domain of azurin 1425

A B PBS Azurin Rusticyanin


0 mM 7 mM

Mel-2

7 mM 14 mM 56 mM
MCF-7

6 mM 12 mM 48 mM
MCF10A1

C
1 20 40 54 60 78 80 100 120 128
Azurin (1JZG_A)
16 140
Auracyanin B (1QHQ_A)
36 155
Rusticyanin (1A3Z)
1 98
Plastocyanin (1IUZ)
1 93
Pseudoazurin(1BQK)

D
Microorganism Accession number

Psae AAG08307 (51) -STAADMQGVVTDGMASGLDKDYLKPDD (77)


Pssy AAO58351 (51) -SKKADASAITTDGMSVGIDKDYVKPDD (77)
Neme AAF41888 (89) IAKAEDMDGVFKDGVGA-ADTDYVKPDD (115)
Vipa NP_800938 (52) -ADTANIQAVGTDGMSAGADNSYVKPDD (78)
Bobr AZBR (51) -TKTADMQAVEKDGIAAGLDNQYLKAGD (77)
Consensus DG D Y K D

Fig. 6. A. Internalization of GSTazu 5077 and its competitive inhibition in the presence of unlabelled cupredoxins. UISO-Mel-2 cells were
incubated with PBS or Alexa fluor 568-conjugated 7 mM GSTazu 5077 at 37C for 1 h (first row). Detailed procedures for competition with
unlabelled proteins are given under Experimental procedures. The same amounts (7 mM) of Alexa fluor-conjugated GSTazu 5077 were mixed
with 7, 14 and 56 mM unlabelled azurin (second row) or with 6, 12 and 48 mM unlabelled rusticyanin (third row). The fusion protein alone or the
mixtures of the fusion proteins with unlabelled cupredoxins were incubated with equal numbers of UISO-Mel-2 cells for 1 h at 37C before the
images were taken.
B. Entry specificity of cupredoxins, azurin and rusticyanin in cancer and normal cells. Alexa fluor 568-conjugated azurin (7 mM) or rusticyanin
(6 mM) was incubated with human melanoma (Mel-2) and breast (MCF-7) cancer and normal mammary epithelial cells (MCF-10A1). Internalization
of cupredoxins was determined by confocal microscopy.
C. Diagram showing the structural alignment of azurin with other cupredoxins, as computed by the VAST algorithm. The N-terminal extended
parts of auracyanin B and rusticyanin, being absent in azurin, have been omitted in the figure. The grey bars indicate the regions of azurin that
can be superimposed on residues from each neighbour. The blank spaces are unaligned regions. The azurin PTD where there is no alignment
with rusticyanin is highlighted by vertical dashed lines. The numbers in parentheses after the names of the cupredoxins are the protein database
accession numbers.
D. Multiple amino acid sequence alignment of the residues comprising the middle part in some known bacterial azurins. Bacterial genus and
species are abbreviated as follows: Psae, Pseudomonas aeruginosa; Pssy, Pseudomonas syringae; Neme, Neisseria meningitidis; Vipa, Vibrio
parahaemolyticus; Bobr, Bordetella bronchiseptica. Numbers of intervening amino acids are given in parenthesis. The CLUSTAL X software (Higgins
and Sharp, 1988) was used to generate this multiple sequence alignment.

10A1 cells (Fig. 6B). Alexa fluor-conjugated rusticyanin, modes of entry and cellular localization in cancer and
however, not only entered the cytosol of UISO-Mel-2 and normal cells.
MCF-7 cancer cells, but also in the normal MCF-10A1 To explore the reasons for the differences in the mode
cells (Fig. 6B). Interestingly, however, unlike in the cancer of entry of these two members of the cupredoxin family
cells where rusticyanin was evenly distributed in the cyto- towards mammalian cells, we checked both the amino
sol, in MCF-10A1 cells, much of the rusticyanin was acid sequence identity and the structural similarities of the
sequestered in the perinuclear space surrounding the azurin PTD with other members of the cupredoxin family.
nucleus. Thus, azurin and rusticyanin show different The sequence identity between azurin and rusticyanin in

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


1426 T. Yamada et al.
the azu 5077 region is less than 20%, as is true of ins in general capable of entering mammalian cells? If
several other cupredoxins (Rienzo et al., 2000; Murphy they do, is there a common mode of entry? Are there
et al., 2002). A structural analysis using VAST algorithm specificities with regard to which cells they enter prefer-
(Gibrat et al., 1996) between azurin and several members entially and why? It is important to note that not only
of the cupredoxin family demonstrated a significant bacterial cupredoxins and cytochromes enter J774-like
similarity between azu 5077 region and that of the mammalian cells, but we have reported that even mam-
cupredoxin auracyanin B, a cupredoxin from the green malian cytochrome c can enter J774 cells when added
thermophilic photosynthetic bacterium Chloroflexus exogenously (Hiraoka et al., 2004). Is the mode of entry
aurantiacus (Bond et al., 2001). Interestingly, however, of mammalian cytochrome c similar to that of bacterial
other members of the cupredoxin family, while demon- cytochrome c? The study reported here is the first step
strating structural similarity with other regions of azurin, towards answering many of these questions. The fact that
lacked this similarity with the azurin PTD (Fig. 6C). azurin and rusticyanin can enter mammalian cells and the
Indeed, when compared with other proteins whose struc- internalization of azurin is significantly impaired at 4C
tures have been deposited in the protein database, there suggests the possible involvement of a receptor-mediated
was very little structural similarity between the azurin PTD endocytic process in azurin internalization. This is con-
and other proteins, demonstrating the unique nature of the firmed by the competition experiments with unlabelled
azurin PTD. It remains to be seen whether the auracyanin azurin and by studies with inhibition by cytochalasin D. Do
B domain, having structural identity with azurin PTD but all cupredoxins use similar receptors for entry? The com-
very little amino acid sequence identity (< 20%), will dem- petition studies indicate that rusticyanin cannot compete
onstrate any entry specificity towards cancer cells. for azurin entry and presumably enters by a different
A lack of structural identity between the azurin PTD and mechanism. Nothing is known about the entry mecha-
other cupredoxins, except auracyanin B, raised an inter- nisms of other cupredoxins that can exert cytotoxicity
esting question: is this PTD conserved in other azurins towards mammalian cells (Yamada et al., 2004c).
found in other bacteria, particularly pathogenic bacteria? An important question that we have to address in future
As the crystal structure of azurins from other pathogenic relates to the specificity of entry. As previously mentioned,
bacteria has not been solved or presented in databases, J774 cells are not normal macrophages but are ascites
we performed a multiple amino acid sequence alignment forms of murine reticulum cell sarcoma (Ralph and
of the residues in the P. aeruginosa PTD region with Nakoinz, 1975). It is interesting to note that azurin exhibits
known sequences of other bacterial azurins from patho- preferential internalization in J774 and human breast can-
gens using the CLUSTAL X alignment program (Higgins and cer MCF-7 cells than the corresponding normal cells such
Sharp, 1988). While the phytopathogenic Pseudomonas as primary peritoneal macrophages and mast cells or
syringae azurin showed high identity with P. aeruginosa mammary epithelial MCF-10F or MCF-10A1 cells. Similar
azurin PTD region (Fig. 6D), an azurin-like protein from selective entry of azurin and induction of apoptosis has
Neisseria meningitidis (Gotschlich and Seiff, 1987; also recently been reported for the human osteosarcoma
Kawula et al., 1987; Cannon, 1989) also showed signifi- cell line U20S but not the normal L-02 cells (Ye et al.,
cant identity with the PTD domain of P. aeruginosa azurin 2005). If azurin entry is indeed due to a receptor-mediated
(Fig. 6D), as did azurins from Vibrio parahaemolyticus endocytic process, then it is likely that such receptors are
and Bordetella bronchiseptica (Fig. 6D). A motif sequence hyperexpressed on the surface of cancer cells than on the
DGX5DX2YXKX2D was found conserved in all surface of normal cells. It would be of great interest to
these azurins. isolate and characterize any putative receptors for azurin
binding and compare their abundance in cancer and nor-
mal cells. The entry domain of azurin can then be used
Discussion
as a vehicle to transport pharmacologically important
Cupredoxins such as azurin and rusticyanin have been compounds preferentially to cancer cells. It remains to be
studied extensively for their electron transfer properties seen whether other cupredoxins or cytochromes will
(Rienzo et al., 2000; Murphy et al., 2002) but there is very exhibit similar specificity for entry into cancer cells for
little information on their ability to enter mammalian cells ultimate use in the treatment of human cancer.
and produce cytotoxic effects. Our recent demonstration An interesting aspect of the present study is our dem-
that the copper-containing redox protein azurin and the onstration of the ability of the azu 5077 peptide to enter
iron (haem)-containing redox protein cytochrome c551 J774 and cancer cells and carry cargo proteins with them,
enter J774 (Yamada et al., 2002a; Hiraoka et al., 2004) thus acting as a PTD. It is not clear from our data whether
and human cancer cells (Yamada et al., 2002b; Punj et al., the azurin PTD may still harbour the cytotoxicity domain.
2004) and induce either apoptosis or inhibition of cell We have recently obtained synthetic peptides correspond-
cycle progression thus raised the question: are cupredox- ing to azu 5077 and demonstrated their cytotoxicity in

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


Entry domain of azurin 1427
J774 and Mel-2 cells (unpubl. obs.). As azurin shows a copy of cytochrome c551 is essential under such condi-
preferential entry towards cancer cells (Figs 1BD and tions (Pearson et al., 2003). An intriguing aspect of the
6B), it could potentially be used as a vehicle to carry cargo azu promoter region is the presence of a CpG island
proteins such as the P. aeruginosa exotoxin A domain III (Fig. 7A) characteristic of those present upstream of many
(PED III) that kills eukaryotic cells by ADP-ribosylating human genes including tumour suppressor genes. The 5
elongation factor 2 and inhibiting protein synthesis (Kreit- CpG islands in the promoter regions of about 60% of
man and Pastan, 1995; Kreitman and Pastan, 2003). PED human genes, including the silenced alleles for selected
III itself cannot enter eukaryotic cells and needs a vehicle imprinted genes and the silenced genes of the inactive
such as the domain II of P. aeruginosa exotoxin A for its X-chromosome of females, which has recently been
entry to the cytosol. Pastan and colleagues have used sequenced, are susceptible to methylation by DNA meth-
various recombinant immunotoxins for targeting CD22, yltransferases that add the methyl groups to the carbon-
CD25 or other cancer-specific targets in order to kill 5 position of cytosine to generate methyl cytosine. Exten-
cancer cells (Kreitman and Pastan, 1995; Kreitman and sive hypermethylation of the CpG islands upstream of
Pastan, 2003). As azurin exerts its cytotoxicity through an many tumour suppressor genes or genes involved in DNA
association with tumour suppressor p53 (Yamada et al., repair or cell cycle, leading to their silencing, is a charac-
2002b; Punj et al., 2004) while PED III exerts its cytotox- teristic of many forms of human cancer (Esteller, 2005;
icity through a KDEL type of sequence in its C-terminus Wang et al., 2005). Whether the presence of a CpG island
for generalized inhibition of protein synthesis, it would be (Fig. 7A), corresponding to Homo sapiens genome
interesting to use a bacterial protein vehicle such as the sequence surrounding the NotI site, clone JNB-106R (Kut-
azurin PTD by fusing it in the N-terminal of PED III to allow senko et al., 2002) with 88% sequence identity (Fig. 7B),
the fusion protein to preferentially enter cancer cells and in the upstream region of the P. aeruginosa azu gene
kill them by a combined cytotoxic mechanism. Such inves- allows its methylation and modulates the expression of
tigations are presently under progress. this gene either during growth in the environment or dur-
The preferential entry of azurin in cancer cells to exert ing P. aeruginosa infection in human remains unknown. It
cytotoxicity raises another interesting question: what is the would be interesting to see whether deletion of this CpG
physiological function of azurin in P. aeruginosa? Azurin- island from the azu promoter region in an in vitro tran-
like proteins are produced by many bacteria such as scription assay will demonstrate any role of this island in
Alcaligenes xylosoxidans, Methylobacillus flagellatum and azu gene expression or whether either prokaryotic or
others (Fig. 6D). The major function of azurin in such eukaryotic DNA methyltransferases will methylate such an
bacteria is believed to be its role in respiration as a com- island to modulate azu gene expression. It is interesting
ponent of the electron transfer chain (Rienzo et al., 2000; to note that this CpG island is unique in P. aeruginosa
Murphy et al., 2002). Although widely believed to be present only in the upstream of the azu gene and is
involved in dissimilatory nitrate reduction in P. aeruginosa, absent in other Pseudomonas genomes and in the
in vivo studies have disproved an obligatory role of azurin genomes of other prokaryotes described in Fig. 6D.
in denitrification and no significant physiological function A second intriguing aspect of azurins physiological
of this cupredoxin has been discerned in P. aeruginosa function, particularly in pathogenic bacteria, is its pre-
(Vijgenboom et al., 1997). Vijgenboom et al. (1997) dem- sumptive role in virulence. P. aeruginosa azurin appears
onstrated that the expression of the azurin gene (azu) was to preferentially attack cancer cells, thus allowing cancer
mediated from two promoters P1 and P2. The proximal regression in vivo. Many bacteria are known to target
promoter P1 was shown to be regulated by ANR, an cancer cells preferentially to normal cells, but how they do
anaerobiosis regulator protein, while the distal promoter it is poorly understood (Sznol et al., 2000; Yu et al., 2004).
P2 had the GG and GC motifs recognized by the alterna- Whether preferential attack to cancer cells by various
tive sigma factor sigma N. These promoters were shown microorganisms (Chakrabarty, 2003; Thomas-Tikhonenko
to be under the control of the stationary-phase sigma and Hunter, 2003) results from elaboration of azurin-like
factor, sigma S, and the function of azurin was believed proteins that might be surface-exposed is also unknown.
to involve electron transfer in response to redox stress, Most interestingly, members of the pathogenic Neisseria
although no effect of azurin was discernible to heat and species including the meningococcus and the gonococ-
cold shock, nutritional shift-down or for growth on various cus produce two outer membrane proteins: a lipoprotein
carbon sources. Thus, the physiological function of azurin called Lip which expresses an outer membrane antigenic
in P. aeruginosa is as yet not clarified. It is interesting to epitope known as H.8 which is common to all pathogenic
note in this context that a mutation in the pseudoazurin Neisseria species; and a second lipoprotein, called Laz.
gene, encoding a cupredoxin also believed to be involved Laz not only contains in its N-terminal a slightly modified
in anaerobic respiration of nitrite in Paracoccus denitrifi- H.8 epitope but also an azurin domain in the C-terminal
cans, has no physiological effect on respiration, although that is highly homologous to P. aeruginosa azurin

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


1428 T. Yamada et al.

A
(-679)cagcagcgggtcgtagaaaccgttcacttcgagcaggcccagcggcttggcgtggtagcccagttggccccaggtccaga

cctcgaacagctcttccagggtgccgagcccgccaggcagggCGatgaaggCGtCGgccagttCGgccatgCGtgccttg

CGCGCGtgcatgcCGtctaccacttccaggcgggtcaggcccttgtggccgatctcggcctcctgcaggctctgcgggat

gatcccgatcacttcgccgccggcggccaatgcggcgtccgccacggtgcccatcagaccgaccgcgccgccaccgtaga

ccagggtcaggccgcgctcggccaggtgccggccgagggccacggcggcttcctggtagaccggggaagcgccggggctg

gcgccacagaatacgcagacggaacgcaaggtcatgatcgactcctgtcgggggtggaaaaaggcgcacagggtagcggc
P2
tgggagcgcttcgaccaagccgtgcgaagcgttgccggacgttgcgtcgcaggcgcgaagcggcacatctgtgctaaaac
P1
aggagTtccccgtagtaaacgccgggcagatcccgctcgatgccccgccacgtccggttcgggtttgacctgaatcagtg

gaactcggtgcccgatcgggcagtctgctctttcaggatTcatcgcccaacctgcctaggaggctgctccATG

B
P.aeruginosa: -576 ggcagggcgatgaaggcgtcggccagttcggccatgcgtgccttgcgcgcgtgcatgccg -517
|||||||||||||| |||||| ||||| | |||||||||| |||||||||||||||||
Homo sapiens: 159 ggcagggcgatgaaagcgtcgctcagttggtgcatgcgtgccatgcgcgcgtgcatgccg 218
Genomic sequence
Accession No. AJ323090

Fig. 7. Analysis of the Pseudomonas aeruginosa azurin promoter region.


A. P1 and P2 promoter regions described by Vijgenboom et al. (1997) are indicated with the transcriptional start site (T) in bold. The DNA region
underlined matched a genomic sequence of Homo sapiens; the CG motifs within this region are indicated in bold capital letters.
B. Sequence alignment (-576 to -517 nucleotides) of the P. aeruginosa azurin upstream region with the H. sapiens genome sequence
demonstrates a very high sequence identity (53/60 nucleotides, 88% identity).

(Gotschlich and Seiff, 1987; Kawula et al., 1987; Cannon, the azurin moiety on the surface (outer membrane) of the
1989). The size of the Lip protein can be somewhat vari- Neisseria species may provide some insights to how bac-
able among the Neisseria species but generally contains teria may target cancer cells preferentially, if the azurin
1120 pentameric AAEAP amino acid repeats, no moiety can be shown to bind cancer cells in preference to
aromatic amino acids and a lipid-modified N-terminal normal cells.
cysteine residue. Lip has been shown to be a potent
inflammatory mediator capable of inducing the release of
Experimental procedures
the chemokine interleukin 8 and the cytokine interleukin
6 as well as activation of the transcription factor NF-kB by Cell cultures
human endocervical epithelial cells and embryonic kidney
The culture conditions of J774 and UISO-Mel-2 cells have been
cells transfected with Toll-like receptor 2 (Fisette et al., described earlier (Yamada et al., 2002a,b; Goto et al., 2003).
2003). The Laz protein, on the other hand, contains imper- Human normal fibroblast cells were cultured in MEM with
fect AAEAP pentameric motifs at the N-terminal but an Eagles salt containing 2 mM L-glutamine, 0.1 mM MEM essen-
127-amino-acid azurin domain at the C-terminal that is tial amino acids and supplemented with 10% heat-inactivated
highly homologous to the P. aeruginosa azurin (Gotschlich fetal bovine serum, 100 units ml-1 penicillin and 100 mg ml-1
and Seiff, 1987; Cannon, 1989). Similar to P. aeruginosa streptomycin. The culturing of MCF-7 and MCF-10F cells has
been described (Punj et al., 2004). Peritoneal macrophages and
azurin, inactivation of the azurin domain in Laz still
mast cells were isolated as described previously (Melnikov
allowed the mutant to grow under both aerobic and anaer- et al., 2000).
obic conditions in the presence of nitrate (Cannon, 1989),
suggesting a non-obligatory role of this moiety in anaero-
bic respiration of Neisseria species. No function of the Laz Plasmid constructions
protein is presently known. It would be interesting to clone
The plasmids expressing fusion GST-truncated azurin derivatives
the azurin gene with or without the gene for the H.8 were constructed by a polymerase chain reaction (PCR) using
epitope and examine their gene products for cytotoxicity proof-reading DNA polymerase. For pGSTazu 36128, an
against various normal and cancer cells. The presence of amplified PCR fragment was introduced into the BamHI and

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


Entry domain of azurin 1429
EcoRI sites of the commercial GST expression vector pGEX5X E. coli BL21 cells were used as the host strain. After induction
(Amersham). The fragment was amplified with pUC19-azu as a with 0.4 mM IPTG at early log phase of growth in L-broth, GST
template and primers, 5-CGGATCC CCG GCA ACC TGC CGA fusion proteins were purified from cell extracts by using Glu-
AGA ACG TCA TGG GC-3 and 5-CGGAATTC GCA TCA CTT tathione Sepharose 4B affinity chromatography and Sephadex
CAG GGT CAG GG-3, where the additionally introduced BamHI 75 gel-filtration column with phosphate-buffered saline (PBS)
and EcoRI sites are underlined, respectively, C-terminus trunca- (Amersham). Purified proteins, WT azurin and GST derivatives
tion of azu gene was cumulatively performed by introducing a or other cupredoxins, labelled with Alexa fluor 568 (Molecular
stop codon using QuickChange site-directed mutagenesis kit Probes), were isolated according to manufacturers instructions.
(Stratagene). For pGSTazu 3650, pGSTazu 3677 and Unbound free fluorescent chemical was removed by gel-filtration
pGSTazu 3689, stop codons were introduced into Ser51, column.
Ser78 and Gly90 respectively. The plasmid carrying pGSTazu
36128 was used as template DNA. Three sets of oligonuclotides
for site-directed mutagenesis are shown as follows. For pGST Azurin internalization in UISO-Mel-2 and fibroblast cells
azu 3650: 5-GGC CAC AAC TGG GTA CTG TGA ACC GCC
GCC GAC ATG CAG-3 and 5-CTG CAT GTC GGC GGC GGT UISO-Mel-2 and fibroblast cells were cultured on coverslips indi-
TCA CAG TAC CCA GTT GTG GCC-3. For pGSTazu 3677: vidually. After overnight incubation, cells on coverslips were
5-CCT GAA GCC CGA CGA CTG ACG TGT CAT CGC CCA washed with fresh media and all three cell lines on coverslips
CAC C-3 and 5-GGT GTG GGC GAT GAC ACG TCA GTC GTC were placed on a culture dish containing 7 mM WT azurin conju-
gated with Alexa fluor 568. The cells were then incubated for
GGG CTT CAG G-3. For pGSTazu 3589: 5-CCA AGC TGA
varying periods at 37C under 5% CO2.
TCG GCT CGT GAG AGA AGG ACT CGG TGA CC-3 and 5-
GGT CAC CGA GTC CTT CTC TCA CGA GCC GAT CAG CTT
GG-3. The plasmid pGSTazu 5077 and pGSTazu 6777
Competition assay
were generated by PCR using pGSTazu 3677 as a template
DNA. Amplified PCR fragments, azu 5077 and azu 6777, were UISO-Mel-2 cells (1 104) were seeded on coverslips and
obtained using forward primer 5-CGGGATCC TGA GCA CCG allowed to grow overnight at 37C. Cells were washed with pre-
CCG CCG ACA TGC AGG G-3 and 5-CGGGATCC CCG GCC warmed fresh media and then incubated with either 7 mM Alexa
TGG ACA AGG ATT ACC TGA AGC CCG-3, where additionally fluor 568-conjugated GSTazu 5077 or a mixture of 7 mM of the
introduced BamHI site is indicated by underlining. The reverse Alexa fluor-conjugated GSTazu 5077 and 7, 14 or 56 mM unla-
primer, 5-CGGAATTC GCA TCA CTT CAG GGT CAG GG-3, belled WT azurin. In separate experiments, competition for the
was utilized in both cases. The plasmid carrying gstazu 5077 entry of 7 mM GSTazu 5077 was examined in the absence or
was used for generating pGSTazu 5066 by introduction of a in the presence of 6, 12 or 48 mM rusticyanin. Controls in the
stop codon in Gly67 using oligonuclotides as follows: 5-GAC absence of Alexa fluor-conjugated GSTazu 5077 or the com-
GGC ATG GCT TCC TGA CTG GAC AAG GAT TAC C-3 and 5- peting proteins were maintained. After 1 h incubation with such
GGT AAT CCT TGT CCA GTC AGG AAG CCA TGC CGT C-3. protein mixture at 37C, cells were washed with PBS to remove
The gfp gene encoding the GFP was also amplified by PCR. excess proteins and nuclei were stained by DAPI. Entry of GST
Forward and reverse primers used were 5-CGGGATCC CCA azu 5077 was observed under confocal microscopy.
TGG TGA GCA AGG GCG-3 and 5-CGGAATTC CTT GTA CAG
CTC GTC CAT GCC G-3 containing BamHI and EcoRI sites at
the 5 end of each oligonuclotides. The resultant PCR fragment Confocal microscopy
was ligated into pGEX5X vector for creating pGSTGFP. For
plasmid DNA carrying gstgfpazu 5077, azu 5077 gene was For preparation of microscope samples, cells were cultured on
amplified by PCR with pGSTazu 5077 as a template and a set coverslips overnight at 37C. Cultured cells were placed at 37C
of primers 5-CCGCTCGAG CCT GAG CAC CGC CGA CAT or 4C for 2 h before protein treatment. Pre-warmed 37C fresh
GCA GGG-3 and 5-TTTTCCTTTTGCGGCCGC TCA GTC GTC media or chilled 4C fresh media were mixed with red fluores-
GGG CTT CAG GTA ATC C-3, where the introduced XhoI and cent-labelled (with Alexa fluor 568) cupredoxins or GST fusion
NotI sites are underlined respectively. Purified azu 5077 frag- derivatives, and incubated with the cells for indicated times. The
ment was introduced into pGSTGFP at XhoI and NotI unique cells were washed with PBS, and fixed with methanol at -20C
restriction enzyme sites. for 5 min. After washing with PBS twice and the addition of
mounting media containing 1.5 mg ml-1 DAPI for staining nuclei
(VECTASHILD, Vector), images were taken by a confocal
Purification of proteins microscope.

Wild-type azurin was purified as described before (Yamada et al.,


2004a). Plasmid DNA, pET9a carrying the rus gene encoding the Azurin internalization in J774 cells as measured by flow
cupredoxin rusticyanin from A. ferrooxidans, was kindly provided cytometry
by Dr Kazuhiko Sasaki, Central Research Institute of Electric
Power Industry, Chiba, Japan. Rusticyanin was isolated from To analyse GSTGFPazu 5077 internalization by flow cytom-
Eschirichia coli BL21 (DE3) harbouring the rus gene as reported etry, 1 105 J774 cells were seeded into six-well plate. After
previously (Sasaki et al., 2003) with some modifications. We overnight incubation, old media were replaced with fresh media
used acetic acid buffer (pH 4.0) and CM-Sepharose (Sigma) and cells were incubated with varying concentrations of GST
instead of b-alanin buffer (pH 4.0) and TSK-gel CM-650 column. GFPazu 5077 at 37C. After GSTGFPazu 5077 treatment
All recombinant GST fusion derivatives were purified as follows: for specified periods, cells were washed with PBS twice, and

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


1430 T. Yamada et al.
collected by FACS (BD FACS Calibur) using CellQuest software similarities in structure comparison. Curr Opin Struct Biol
(Becton Dickinson). 6: 377385.
Goto, M., Yamada, T., Kimbara, K., Horner, J., Newcomb, M.,
Das Gupta, T.K., and Chakrabarty, A.M. (2003) Induction
of apoptosis in macrophages by Pseudomonas aeruginosa
Structural similarity between azurin and other cupredoxins
azurin: tumour-suppressor protein p53 and reactive oxygen
The crystal structures of azurin from P. aeruginosa (1JZG_A), species, but not redox activity, as critical elements in cyto-
auracyanin B from C. aurantiacus (1QHQ_A), rusticyanin from toxicity. Mol Microbiol 47: 549559.
A. ferrooxidans (1A3Z), plastocyanin from Ulva pertusa (1IUZ)
Gotschlich, E.C., and Seiff, M.E. (1987) Identification and
gene structure of an azurin-like protein with a lipoprotein
and pseudoazurin from Achromobacter cycloclastes (1BQK), as
signal peptide in Neisseria gonorrhoeae. FEMS Microbiol
shown in Fig. 6C, were aligned using the VAST algorithm (Gibrat
Lett 43: 253255.
et al., 1996), available at http://www.ncbi.nlm.nih.gov/Structure/.
Gough, J., and Chothia, C. (2004) The linked conservation
The amino acid sequence alignment displayed in Fig. 6D was of structure and function in a family of high diversity: the
generated using CLUSTAL X (Higgins and Sharp, 1988). monomeric cupredoxins. Structure 12: 917925.
Grossmann, T.G., Hall, J.F., Kanbi, L.D., and Hasnain, S.S.
(2002) The N-terminal extension of rusticyanin is not
Acknowledgements responsible for its acid stability. Biochemistry 41: 3613
3619.
This investigation was supported by a Public Health Service Hallbrink, M., Floren, A., Elmquist, A., Pooga, M., Barfai, T.,
Grant ES-04050-19 from the National Institute of Environmental and Langel, U. (2001) Cargo delivery kinetics of cell-
Health Sciences. A.M.F. acknowledges sabbatical grants from penetrating peptides. Biochim Biophys Acta 1515: 101
Fundacao para Ciencia e Tecnologia (FCT) BSAB-457, Portugal, 109.
and Fundacao Calouste Gulbenkian. Higgins, D.G., and Sharp, P.M. (1988) CLUSTAL: a package
for performing multiple sequence alignment on a micro-
computer. Gene 73: 237244.
References Hiraoka, Y., Yamada, T., Goto, M., Das Gupta, T.K., and
Chakrabarty, A.M. (2004) Modulation of mammalian cell
Appia-Ayme, C., Guiliani, N., Ratouchniak, J., and Bonnefoy, growth and death by prokaryotic and eukaryotic cyto-
V. (1999) Characterization of an operon encoding two c- chrome c. Proc Natl Acad Sci USA 101: 64276432.
type cytochromes, and aa(3)-type cytochrome oxidase, Kawula, T.H., Spinola, S.M., Klapper, D.G., and Cannon, J.G.
and rusticyanin in Thiobacillus ferrooxidans ATCC 33020. (1987) Localization of a conserved epitope and an azurin-
Appl Environ Microbiol 65: 47814787. like domain in the H.8 protein of pathogenic Neisseria. Mol
Bond, C.S., Blankenship, R.E., Freeman, H.C., Guss, J.M., Microbiol 1: 179185.
Maher, M.J., Selvaraj, F.M., et al. (2001) Crystal structure Kreitman, R.J., and Pastan, I. (1995) Importance of the
of auracyanin, a blue copper protein from the green ther- glutamate residue of KDEL in increasing the cytotoxicity of
mophilic photosynthetic bacterium Chloroflexus aurantia- Pseudomonas exotoxin derivatives and for increased bind-
cus. J Mol Biol 306: 4767. ing to the KDEL receptor. Biochem J 307: 2937.
Cannon, J.G. (1989) Conserved lipoproteins of pathogenic Kreitman, R.J., and Pastan, I. (2003) Immunobiological treat-
Neisseria species bearing the H.8 epitope: lipid-modified ments of hairy-cell leukemia. Best Pract Res Clin Haematol
azurin and H.8 outer membrane protein. Clin Microbiol Rev 16: 117133.
2: S1S4. Kutsenko, A.S., Gizatullin, R.Z., Al-Amin, A.N., Wang, F.,
Chakrabarty, A.M. (2003) Microorganisms and cancer: quest Kvasha, S.M., Podowski, R.M., et al. (2002) NotI flanking
for a therapy. J Bacteriol 185: 26832686. sequences: a tool for gene discovery and verification of the
Chaloin, L., Bigeye, P., Loup, C., Marin, M., Galled, N., human genome. Nucleic Acids Res 30: 31633170.
Piechaczyk, M., et al. (2001) Improvement of porphyrin Lindgren, M., Hallbrink, M., Prochiantz, A., and Langel, U.
cellular delivery and activity by conjugation to a carrier (2000) Cell-penetrating peptides. Trends Pharmacol Sci
peptide. Bioconjug Chem 12: 691700. 21: 99103.
Elliott, G., and OHare, P. (1997) Intercellular trafficking and Lippincott Schwartz, J., Donaldson, J.G., Schweizer, A.,
protein delivery by a herpesvirus structural protein. Cell 88: Berger, E.G., Hauri, H.P., Yuan, I.C., and Klausner, R.D.
223233. (1990) Microtubule-dependent retrograde transport of pro-
Esteller, M. (2005) DNA methylation and cancer therapy: new teins into the ER in the presence of brefeldin A suggests
developments and expectations. Curr Opin Oncol 17: 55 an ER recycling pathway. Cell 60: 821836.
60. Liu, Y., Jones, M., Hingtgen, C.M., Bu, G., Laribee, N., Tanzi,
Fisette, P.L., Ram, S., Andersen, J.M., Guo, W., and Ingalls, R.E., et al. (2000) Uptake of HIV-1 tat protein mediated by
R.R. (2003) The Lip lipoprotein from Neisseria gonor- low-density lipoprotein receptor-related protein disrupts the
rhoeae stimulates cytokine release and NF-kB activation in neuronal metabolic balance of the receptor ligands. Nat
epithelial cells in a Toll-like receptor 2-dependent manner. Med 6: 13801387.
J Biol Chem 278: 4625246260. Melnikov, A., Zaborina, O., Dhiman, N., Prabhakar, B.S.,
Fittipaldi, A., Ferari, A., Zoppe, M., Arcangeli, C., Pelligrini, Chakrabarty, A.M., and Hendrickson, W. (2000) Clinical
V., Beltram, F., and Giacca, M. (2003) Cell membrane lipid and environmental isolates of Burkholderia cepacia exhibit
rafts mediate caveolar endocytosis of HIV-1 Tat fusion pro- differential cytotoxicity towards macrophages and mast
teins. J Biol Chem 278: 3414134149. cells. Mol Microbiol 36: 14811493.
Gibrat, J.F., Madej, T., and Bryant, S.H. (1996) Surprising Morris, M.C., Depollier, J., Mery, J., Heitz, F., and Divita, G.

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431


Entry domain of azurin 1431
(2001) A peptide carrier for the delivery of biologically Tyagi, M., Rusnati, M., Presta, M., and Giacca, M. (2001)
active proteins into mammalian cells. Nat Biotechnol 19: Internalization of HIV-1 tat requires cell surface hepa-
11731176. ran sulfate proteoglycans. J Biol Chem 276: 3254
Murphy, L.M., Dodd, F.E., Yousafzai, F.K., Eady, R.R., and 3261.
Hasnain, S.S. (2002) Electron donation between copper Vijgenboom, E., Busch, J.E., and Canters, G.W. (1997) In
containing nitrite reductases and cupredoxins: the nature vivo studies disprove an obligatory role of azurin in denitri-
of proteinprotein interaction in complex formation. J Mol fication in Pseudomonas aeruginosa and show that azu
Biol 315: 859871. expression is under control of RpoS and ANR. Microbiol-
Pearson, I.V., Page, M.D., van Spannig, R.J.M., and Fergu- ogy 143: 28532863.
son, S.J. (2003) A mutant of Paracoccus denitrificans with Wang, W., Huper, G., Guo, Y., Murphy, S.K., Olson,J.A.,
disrupted genes coding for cytochrome c550 and pseudoa- Jr, and Marks, J.R. (2005) Analysis of methylation
zurin establishes these two proteins as the in vivo electron sensitive transcription identifies GADD45a as a frequently
donors to cytochrome cd1 nitrite reductase. J Bacteriol 185: methylated gene in breast cancer. Oncogene 24: 2705
63086315. 2714.
Punj, V., Bhattacharyya, S., Saint-Dic, D., Vasu, C., Cunning- Yamada, T., Goto, M., Punj, V., Zaborina, O., Kimbara, K.,
ham, E.A., Graves, J., et al. (2004) Bacterial cupredoxin Das Gupta, T.K., and Chakrabarty, A.M. (2002a) The bac-
azurin as an inducer of apoptosis and regression in human terial redox protein azurin induces apoptosis in J774 mac-
breast cancer. Oncogene 23: 23672378. rophages through complex formation and stabilization of
Ralph, P., and Nakoinz, I. (1975) Phagocytosis and cytolysis the tumor suppressor protein p53. Infect Immun 70: 7054
by a macrophage tumour and its cloned cell line. Nature 7062.
257: 393394. Yamada, T., Goto, M., Punj, V., Zaborina, O., Chen, M.L.,
Rienzo, F.D., Gabdoulline, R.R., Menziani, M.C., and Wade, Kimbara, K., et al. (2002b) Bacterial redox protein azurin,
R.C. (2000) Blue copper proteins: a comparative analysis tumor suppressor protein p53, and regression of cancer.
of their molecular interaction properties. Protein Sci 9: Proc Natl Acad Sci USA 99: 1409814103.
14391454. Yamada, T., Hiraoka, Y., Ikehata, M., Kimbara, K., Avner,
Sasaki, K., Ida, C., Ando, A., Matsumoto, N., Saiki, H., and B.S., Das Gupta, T.K., and Chakrabarty, A.M. (2004a)
Ohmura, N. (2003) Respiratory isozyme, two types of rus- Apoptosis or growth arrest: modulation of tumor suppres-
ticyanin of Acidithiobacillus ferrooxidans. Biosci Biotechnol sor p53s specificity by bacterial redox protein azurin. Proc
Biochem 67: 10391047. Natl Acad Sci USA 101: 47704775.
Schwarze, S.R., Ho, A., Vocero-Akbani, A., and Dowdy, S.F. Yamada, T., Hiraoka, Y., Das Gupta, T.K., and Chakrabarty,
(1999) In vivo protein transduction: delivery of a biologically A.M. (2004b) Rusticyanin, a bacterial electron transfer pro-
active protein into the mouse. Science 285: 15691572. tein, causes G1 arrest in J774 and apoptosis in human
Schwarze, S.R., Hruska, K.A., and Dowdy, S.F. (2000) Pro- cancer cells. Cell Cycle 3: 11821187.
tein transduction: unrestricted delivery into all cells? Trends Yamada, T., Hiraoka, Y., Das Gupta, T.K., and Chakrabarty,
Cell Biol 10: 290295. A.M. (2004c) Regulation of mammalian cell growth and
Snyder, E.L., Meade, B.R., Saenz, C.C., and Dowdy, S.F. death by bacterial redox proteins: relevance to ecology and
(2004) Treatment of terminal peritoneal carcinomatosis by cancer therapy. Cell Cycle 3: 752755.
a transducible p53-activating peptide. PLoS Biol 2: 186 Ye, Z.M., Miao, X.D., Yang, D.S., Xu, R.Z., Huang, X., and
193. Ge, F.F. (2005) Selective inducement effect of bacterial
Sofer, A., and Futerman, A.H. (1995) Cationic amphiphilic redox protein azurin on apoptosis of human osteosarcoma
drugs inhibit the internalization of cholera toxin to the Golgi cell line U20S. Ai Zheng 24: 298304 (in Chinese).
apparatus and the subsequent elevation of cyclic AMP. Yu, Y.A., Shabahang, S., Timiryasova, T.M., Zhang, Q.,
J Biol Chem 270: 1211712122. Beltz, R., Gentschev, I., et al. (2004) Visualization of tumors
Suzuki, T., Futaki, S., Niwa, M., Tanaka, S., Ueda, K., and and metastases in live animals with bacteria and vaccinia
Sugiura, Y. (2002) Possible existence of common internal- virus encoding light-emitting proteins. Nat Biotechnol 22:
ization mechanisms among arginine-rich peptides. J Biol 313320.
Chem 277: 24372443. Zaborina, O., Dhiman, N., Chen, M.L., Kostal, J., Holder, I.A.,
Sznol, M., Lin, S.L., Bermudes, D., Zheng, L.-M., and King, and Chakrabarty, A.M. (2000) Secreted products of a
I. (2000) Use of preferentially replicating bacteria for the nonmucoid Pseudomonas aeruginosa strain induce two
treatment of cancer. J Clin Invest 105: 10271030. modes of macrophage killing: external-ATP-dependent,
Thomas-Tikhonenko, A., and Hunter, C.A. (2003) Infection P2Z-receptor-mediated necrosis and ATP-independent,
and cancer: the common vein. Cytokine Growth Factor Rev caspase-mediated apoptosis. Microbiology 146: 2521
14: 6777. 2530.

2005 Blackwell Publishing Ltd, Cellular Microbiology, 7, 14181431

Das könnte Ihnen auch gefallen