You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/232969231

Protein metalation by metal-based drugs: Reactions of cytotoxic gold


compounds with cytochrome c and lysozyme

Article in European Journal of Biochemistry · November 2012


DOI: 10.1007/s00775-012-0952-6 · Source: PubMed

CITATIONS READS

39 262

7 authors, including:

Chiara Gabbiani Lara Massai


Università di Pisa University of Florence
138 PUBLICATIONS 5,617 CITATIONS 59 PUBLICATIONS 946 CITATIONS

SEE PROFILE SEE PROFILE

Elena Michelucci Laura Maiore


Italian National Research Council Università degli Studi di Sassari
48 PUBLICATIONS 1,020 CITATIONS 27 PUBLICATIONS 555 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Electrochemical and spectroelectochemical behavior of organometallic compounds in configuration d8 View project

Arsenoplatin-1 is a Dual Pharmacophore Anti-Cancer Agent View project

All content following this page was uploaded by Laura Maiore on 19 May 2014.

The user has requested enhancement of the downloaded file.


J Biol Inorg Chem (2012) 17:1293–1302
DOI 10.1007/s00775-012-0952-6

ORIGINAL PAPER

Protein metalation by metal-based drugs: reactions of cytotoxic


gold compounds with cytochrome c and lysozyme
Chiara Gabbiani • Lara Massai • Federica Scaletti •
Elena Michelucci • Laura Maiore • Maria Agostina Cinellu •

Luigi Messori

Received: 6 August 2012 / Accepted: 7 October 2012 / Published online: 7 November 2012
Ó SBIC 2012

Abstract Protein metalation processes are crucial for the lysozyme, through UV–visible absorption spectroscopy
mechanism of action of several anticancer metallodrugs and electrospray ionisation mass spectrometry (ESI MS) to
and warrant deeper characterisation. We have explored the characterise the inherent protein metalation processes.
reactions of three cytotoxic gold(III) compounds—namely Notably, Auoxo6 and Au2phen produced stable protein
[(bipy2Me)2Au2(l-O)2][PF6]2 (where bipy2Me is 6,60 -dime- adducts where one or more ‘‘naked’’ gold(I) ions are pro-
thyl-2,20 -bipyridine) (Auoxo6), [(phen2Me)2Au2(l-O)2] tein-coordinated; very characteristic is the case of cyto-
[PF6]2 (where phen2Me is 2,9-dimethyl-1,10-phenanthro- chrome c, which upon reaction with Auoxo6 or Au2phen
line) (Au2phen) and [(bipydmb-H)Au(OH)][PF6] [where preferentially forms ‘‘tetragold’’ adducts with four protein-
bipydmb-H is deprotonated 6-(1,1-dimethylbenzyl)-2,20 - bound gold(I) ions. In turn, Aubipyc afforded monometa-
bipyridine] (Aubipyc)—with two representative model lated protein adducts where the structural core of the
proteins, i.e. horse heart cytochrome c and hen egg white gold(III) centre and its ?3 oxidation state are conserved.
Auranofin yielded protein derivatives containing the intact
auranofin molecule. Additional studies were performed to
Electronic supplementary material The online version of this
article (doi:10.1007/s00775-012-0952-6) contains supplementary assess the role played by a reducing environment in protein
material, which is available to authorized users. metalation. Overall, the approach adopted provides
detailed insight into the formation of metallodrug–protein
C. Gabbiani
derivatives and permits trends, peculiarities and mecha-
Department of Chemistry and Industrial Chemistry,
University of Pisa, nistic details of the underlying processes to be highlighted.
Via Risorgimento 35, In this respect, electrospray ionisation mass spectrometry is
56126 Pisa, Italy a very straightforward and informative research tool. The
protein metalation processes investigated critically depend
L. Massai  F. Scaletti  L. Messori (&)
Department of Chemistry, on the nature of both the metal compound and the inter-
University of Florence, acting protein and also on the solution conditions used;
Via della Lastruccia 3, thus, predicting with accuracy the nature and the amounts
50019 Sesto Fiorentino, Italy
of the adducts formed for a given metallodrug–protein pair
e-mail: luigi.messori@unifi.it
is currently extremely difficult.
E. Michelucci
Mass Spectrometry Centre (CISM), Keywords Anticancer drugs  Proteins  Gold compounds 
University of Florence,
Mechanism of action  Mass spectrometry
Via U. Schiff 6,
50019 Sesto Fiorentino, Italy

L. Maiore  M. A. Cinellu Introduction


Department of Chemistry and Pharmacy,
University of Sassari,
Via Vienna 2, A few recent studies highlighted the importance of gold
07100 Sassari, Italy compounds as a new family of cytotoxic agents with the

123
1294 J Biol Inorg Chem (2012) 17:1293–1302

potential of becoming effective anticancer drug candidates co-workers [15, 16] concerning serum albumin and cy-
[1–3]. Indeed, a variety of gold compounds, either gold(III) clophilin. Talib et al. [17] used mass spectrometry (MS)
or gold(I), bearing different structural motifs were reported methods to investigate the binding of various gold(I) com-
to cause effective cell death in vitro [4–7] in numerous pounds to serum albumin. A systematic study conducted in
cancer cell lines; for a few gold(III) compounds, i.e. our laboratory a few years ago considered the reactions of
gold(III) porphyrins and gold(III) dithiocarbamates, pre- various gold compounds with serum albumin and investi-
liminary but very promising in vivo results were also gated the underlying interaction mechanisms [9, 18, 19].
obtained [1, 8]. Very recent studies, mainly based on electrospray ionisa-
The modes of action of antiproliferative gold com- tion (ESI) MS, analysed the reactions of cytotoxic gold
pounds are still largely unknown and a matter of intense compounds with cytochrome c and lysozyme and descri-
research and debate. They appear to be multifaceted and bed the resulting adducts [20, 21].
deeply distinct from those of clinically established, DNA- During the last few years, we have prepared and char-
damaging platinum compounds. Notably, there is a grow- acterised three novel gold(III) complexes—namely
ing body of evidence suggesting that some selected protein [(bipy2Me)2Au2(l-O)2][PF6]2 (where bipy2Me is 6,60 -dime-
targets rather than nucleic acids primarily mediate the thyl-2,20 -bipyridine) (Auoxo6), [(phen2Me)2Au2
2Me
biological effects of cytotoxic gold compounds [9, 10]. A (l-O)2][PF6]2 (where phen is 2,9-dimethyl-1,10-phe-
few studies postulated that the cytotoxic effects of gold nanthroline) (Au2phen) and [(bipydmb-H)Au(OH)][PF6]
compounds are driven by a specific antimitochondrial (where bipydmb-H is deprotonated 6-(1,1-dimethylbenzyl)-
mechanism, ultimately leading to apoptotic cancer cell 2,20 -bipyridine) (Aubipyc)—that revealed quite promising
death [11–13]; however, this latter hypothesis still requires antiproliferative properties when tested in vitro on a variety
conclusive validation. of cancer cell lines. The three complexes mentioned are
Over the last 30 years, a number of studies have shown in Fig. 1; the schematic structure of auranofin, a
appeared concerning the reactions of gold compounds with clinically established gold(I) drug used here for compari-
proteins, analysed at the molecular level. Some pioneering son purposes, is also reported.
work was conducted by Frank Shaw and co-workers [14] Auoxo6 and Au2phen are dioxo-bridged dinuclear
into the reactions of auranofin with serum albumin. Other gold(III) complexes with 6,60 - dimethyl-2,20 -bipyridyne
important studies were reported by Peter Sadler and and 2,9-dimethyl-1,10-phenanthroline, respectively, as

Fig. 1 Structures of the various


gold compounds considered in
this study: a auranofin
(Mr 678.50); b Auoxo6
(Mr 1,084.41); c Au2phen
(Mr 1,132,39); d Aubipyc
(Mr 634,32)
a

b c

123
J Biol Inorg Chem (2012) 17:1293–1302 1295

ancillary ligands [22–24]. Both gold(III) centres in Auoxo6 possesses at least four strong anchoring sites for metals; in
and Au2phen display a classic square-planar coordination, contrast, lysozyme manifested a lower tendency to form
with slight square-pyramidal distortions. Aubipyc is a platinum adducts, and typically yielded monometalated
gold(III) cyclometalated derivative of 6-(1,1-dimethylben- platinum derivatives through platinum coordination to its
zyl)-2,20 -bipyridine, which features an N,N,C sequence of unique histidine residue, i.e. His-15.
donor atoms of the terdentate bipyridine ligand and an The main goal of the present study concerning interac-
oxygen atom of a hydroxo ligand coordinated to the tions of a few gold-based drugs with proteins is to identify
gold(III) centre in a square-planar geometry [25]. Auran- and define, at the molecular level, the modes of protein
ofin, 2,3,4,6-tetra-O-acetyl-1-thio-b-D-pyranosato-S-(tri- metalation by these gold compounds. Metalation is the
ethylphosphine)gold(I), is a linear gold(I) complex of the process through which proteins are modified upon reaction
type R3P-Au(I)-SR0 , where SR0 is a sugar thiolate. with metal-containing agents; such reactions typically
The cytotoxic properties of the above-mentioned involve formation of adducts between the protein and a
gold(III) complexes were evaluated in vitro on a standard metal species (often a molecular fragment derived from the
36 cancer cell line panel available in Oncotest (Freiburg, starting metal compound). The metallic fragment may be
Germany) according to established procedures [26, 27]. bound to the protein either by a direct coordinative bond or
The COMPARE algorithm [28, 29] applied to the analysis through a non-covalent interaction. Precise information on
of the growth inhibition data obtained revealed that the the actual metalation mechanisms may be achieved quite
profiles of Au2phen are very similar to those of Auoxo6; realistically through a detailed structural characterisation of
this behaviour reflects the pronounced structural analogy metal–protein adducts including adduct quantification with
between these two compounds [22]. Tentatively, the patterns respect to native unmodified proteins, identification of the
of antiproliferative activity obtained for Au2phen and Auoxo6 nature and the number of protein-bound metallic fragments,
were referred to inhibition of histone deacetylase on the basis and localisation of the anchoring sites for metallic fragments.
of bioinformatic analysis. At variance, Aubipyc, although ESI MS appears to be a very appropriate and powerful tool to
being appreciably cytotoxic to A2780 ovarian cancer cells, gain this kind of information; conversely, absorption spec-
was only moderately effective in the Oncotest panel. Auran- troscopy offers the opportunity to monitor continuously, in
ofin was confirmed to be highly cytotoxic in the Oncotest ‘‘real time’’, the various metallodrug–protein species.
panel, with a mode of action resembling that of typical anti-
proteasomal agents [8]. As mentioned above, correlations in
the antiproliferative profiles between the various gold com- Materials and methods
pounds and cisplatin were very poor, implying the occurrence
of drastically different modes of action. Metal complexes and proteins
On the whole, COMPARE analysis of these compounds
as well as of several other cytotoxic gold compounds Au2phen [21], Auoxo6 [6] and Aubipyc [25] were syn-
suggested that a variety of proteins such as histone thesised and characterised as described previously (see the
deacetylase, cyclin-dependent kinase, proteasome proteins respective references). Auranofin was purchased from
and mammalian target of rapamycin might be reliable Vinci-Biochem. Horse heart cytochrome c (C7752) and
biomolecular targets and thus account for their biological chicken hen egg white lysozyme (L7651) were purchased
effects; in other words, the biological actions of the gold from Sigma and used as received.
compounds investigated might be best interpreted in terms
of metalation and inactivation of a few crucial intracellular Physicochemical measurements
proteins that are effective cancer targets.
The great current interest in interactions between metal- UV–vis spectra were recorded with a Varian Cary 50
based drugs and proteins prompted us to conduct a sys- UV–vis spectrophotometer. Mass spectra were recorded
tematic analysis of the reactions occurring between the with an LTQ Orbitrap high-resolution mass spectrometer
above-mentioned gold compounds and two model proteins, (Thermo Scientific, San Jose, CA, USA) equipped with a
namely horse heart cytochrome c and hen egg white conventional ESI source.
lysozyme. These small proteins are very amenable to ESI
MS analysis and were specifically chosen for the present UV–vis spectrophotometric studies
investigation. The interactions of these two proteins with a
variety of metalbased drugs, and in particular with cis- Small amounts of freshly prepared concentrated solutions
platin, were previously investigated in detail [30, 31]. of the individual complexes in dimethyl sulfoxide (or
Cytochrome c was found to produce several platinum EtOH/H2O for auranofin) were diluted in the reference
adducts with various metal-to-protein stoichiometries as it buffer (10 mM phosphate, pH 7.4). The concentration of

123
1296 J Biol Inorg Chem (2012) 17:1293–1302

each gold compound in the final sample was 10-5 M. The


resulting solutions were monitored by collection of the
electronic spectra for 24 h at room temperature.

Interactions with proteins

Electronic spectra of the model protein (lysozyme or


cytochrome c) at 10-5 M were recorded before and after
the addition of each gold complex at a stoichiometric ratio
of 3:1 (metal to protein) for 24 h at room temperature in
10 mM phosphate buffer, pH 7.4. Similar spectrophoto-
metric studies were conducted in the presence of a reducing
Fig. 2 Time course UV–vis spectra of Auoxo6 dissolved in 10 mM
agent, i.e. glutathione (GSH) or ascorbic acid, added to the phosphate buffer, pH 7.4, before and after addition of 1 mM
various reaction batches to a final concentration of 1 mM. glutathione. The concentration of the complex was 10-5M. The
spectra were recorded before (red line) and after the addition of the
reducing agent at 0 min (blue line), 5 min (green line), 10 min (violet
Sample preparation and MS analysis
line), 15 min (grey line) and 1 h (light-blue line)

Metal complex–protein adducts were prepared by mixing


equivalent amounts of the three proteins (10-4 M) in obtained for the same compounds under slightly different
25 mM tetramethylammonium acetate buffer, pH 7.4. experimental conditions [6, 25]. It is remarkable that the
Then, the three gold(III) complexes and auranofin were ?3 oxidation state is essentially conserved in all cases over
added (3:1 metal-to-protein ratio) to the solution and the the whole observation period.
mixture was incubated at 37 °C for 72 h. After a 20-fold In contrast, important spectral changes were highlighted
dilution with water, ESI MS spectra were recorded by when these gold(III) complexes were challenged with an
direct introduction of the sample at a flow rate of 5 ll/min excess of two biologically occurring reducing agents, i.e.
into an Orbitrap high-resolution mass spectrometer ascorbic acid and GSH (Fig. S2). Auoxo6 and Au2phen
(Thermo Scientific, San Jose, CA, USA) equipped with a underwent quick and facile reduction upon addition of an
conventional ESI source. The working conditions were as excess of either reducing agent, which was documented by
follows: spray voltage 3.1 kV, capillary voltage 45 V and evident spectral changes. The reaction of Auoxo6 with
capillary temperature 220 °C. The sheath and the auxiliary GSH is very representative as shown in Fig. 2; indeed, the
gases were set at 17 (arbitrary units) and 1 (arbitrary unit), ligand-to-metal charge transfer band centred at 325 nm
respectively,. For acquisition, Xcalibur 2.0 (Thermo Sci- rapidly disappears upon addition of GSH. Conversely, in
entific) was used and monoisotopic and average deconvo- the case of Au2phen, we noticed large changes in the shape
luted masses were obtained by using the integrated Xtract and intensity of the composite band centred at 280 nm that
tool. For spectrum acquisition, a nominal resolution (at m/ are suggestive of reduction of gold(III) (Fig. S2). Aubipyc
z 400) of 100,000 was used. was far more resistant to reduction: indeed, ascorbic acid
failed to reduce it, whereas an excess of GSH induced a
progressive–although slow—reduction of the gold(III)
Results centre (Fig. S2).

Solution behaviour of the three gold(III) compounds Reactions of gold compounds with model proteins:
spectrophotometric analysis
Before studying their interactions with proteins, we ana-
lysed the solution behaviour of the three gold(III) com- The interactions of these gold(III) compounds with the two
plexes under well-controlled experimental conditions. aforementioned model proteins were explored. Each of the
UV–vis absorption spectroscopy was selected as the gold(III) compounds was reacted with cytochrome c or
method to monitor continuously the behaviour of their lysozyme at a standard metal-to-protein molar ratio of 3:1
gold(III) chromophores in the reference buffer (10 mM according to previously defined experimental procedures
phosphate buffer, pH 7.4). All three gold(III) compounds and the resulting solutions were incubated for 72 h at
manifested appreciable stability when monitored for 24 h 37 °C (25 mM tetramethylammonium acetate buffer, pH
at 20 °C, as documented by the substantial invariance of 7.4). Samples were continuously monitored through spec-
their absorption spectra (see Fig. S1). These observations trophotometric analysis. For comparison purposes, similar
are in agreement with previous spectrophotometric results experiments were conducted using auranofin as the

123
J Biol Inorg Chem (2012) 17:1293–1302 1297

metalating agent. At the end of the incubation period, ESI MS spectra of metallodrug–protein samples
samples were analysed by ESI MS as described in the next
section. ESI MS is a very powerful analytical tool to characterise
UV–vis spectrophotometric analysis of metallodrug– metallodrug–protein interactions at the molecular level [32,
protein samples allows the continuous monitoring of the 33]. Accordingly, ESI MS analysis of the previously
various gold(III) centres in the presence of these model described samples allowed us to identify and characterise a
proteins. In the case of cytochrome c—i.e. a metalloprotein number of metal–protein adducts whose presence is hardly
characterised by intense transitions in the visible region—it documented in the electronic absorption spectra owing to
is possible to monitor simultaneously both the metallo- the intrinsic limitations of that technique. In particular, ESI
drug’s chromophore and the active site of the protein in the MS measurements permitted the nature of protein-bound
course of their interaction. metallic fragments to be disclosed and their binding stoi-
From spectral inspection, it is apparent that addition of chiometries to be determined, thus providing indirect
protein does not affect the behaviour of the various mechanistic insight into the respective metalation pro-
gold(III) chromophores (Fig. 3). Yet, modest—but slowly cesses. As a variety of different situations were encoun-
progressive—spectral changes were detected in the case of tered depending on the nature of the metallodrug and, to a
the reaction of Auoxo6 with cytochrome c, as shown in lesser extent, of the protein, the various cases will be
Fig. 3. Some significant changes in the shape of the com- illustrated separately.
posite band of Au2phen centred at 280 nm were also
detected that are suggestive of the occurrence of partial Auoxo6 and Au2phen
reduction of gold(III). Conversely, the spectra of the vari-
ous metallodrug–cytochrome c systems reveal that the These two gold complexes are described together as they
protein chromophore is substantially stable for 24 h, with manifested rather similar reactivity patterns with regard to
cytochrome c remaining in its oxidised ferric form. both lysozyme and cytochrome c. ESI MS spectra of

Fig. 3 Time course UV–vis


spectra of gold compounds
dissolved in 10 mM phosphate
buffer, pH 7.4, upon 24 h
incubation with lysozyme or
cytochrome c. The protein
concentration was 10-5 M and
the metallodrug-to-protein
molar ratio was 3:1. The plots
on the left show the spectral
features of cytochrome c upon
addition of Au2phen (a),
Auoxo6 (b) and Aubipyc (c).
The plots on the right show the
spectral features of lysozyme
upon addition of Au2phen (d),
Auoxo6 (e) and Aubipyc (f). All
reported spectra were recorded
before (red line) and after the
addition of the three gold
compounds at 0 h (green line),
1 h (violet line), 12 h (blue line)
and 24 h (grey line)

123
1298 J Biol Inorg Chem (2012) 17:1293–1302

Fig. 4 LTQ Orbitrap electrospray ionisation (ESI) mass spectra of presence of cytochrome c (A) or lysozyme (B). The plots on the right
gold compounds dissolved in 25 mM tetramethylammonium acetate show Auoxo6 in the presence of cytochrome c (C) or lysozyme (D).
(TMeAmAc) buffer, pH 7.4, in the presence of the two representative All spectra show the peak of the native protein (a) and of gold(I)–
proteins (lysozyme and cytochrome c) after 72 h incubation at 37 °C. protein adducts with stoichiometries of 1:1 (b), 2:1 (c), 3:1 (d), 4:1
The protein concentration was 10-4 M (with a metal complex to (e) and 5:1 (f)
protein molar ratio of 3:1). The plots on the left show Au2phen in the

protein solutions incubated with these two compounds for position of the peaks permits us to assign these adducts to
72 h at a 3:1 metallodrug-to-protein molar ratio are shown one or more ‘‘naked’’ gold(I) ions binding to protein; no
comparatively in Fig. 4. evidence of ‘‘mass shifts’’ corresponding to the original
From careful inspection of this Fig. 4, it is evident that a gold(III) ligands was obtained, implying that reduction of
number of metal–protein adducts are formed in the various gold(III) and complex disruption always precede protein
cases, although in greatly different amounts, as can be binding. Very interestingly, cytochrome c favours the for-
judged by comparing the relative peak intensities. Au2phen mation of a metal protein adduct with a stoichiometry of
manifests a very high reactivity with cytochrome c, which 4:1, i.e. four gold(I) ions bound to the protein (see the peak
is accompanied by the formation of relatively large with molecular mass of 13,142 Da); this observation is in
amounts of adducts; in the other cases, only small amounts agreement with previous studies on the reaction of cyto-
of metal–protein adducts are formed. In all cases, the chrome c with gold saccharinate compounds [20] and with

123
J Biol Inorg Chem (2012) 17:1293–1302 1299

Fig. 5 LTQ Orbitrap ESI mass spectra of Aubipyc dissolved in ratio of 3:1). Both spectra show peaks corresponding to the native
25 mM TMeAmAc buffer, pH 7.4, in the presence of lysozyme protein (a) and the [(bipydmb-H)Au] moiety–protein adduct [where
(A) and cytochrome c (B) after 72 h incubation at 37 °C. The protein bipydmb-H is deprotonated 6-(1,1-dimethylbenzyl)-2,20 -bipyridine; Mr
concentration was 10-4 M (with a metal complex to protein molar 472.36] (b)

the presence of four major anchoring sites for metals on the probably by non-covalent interaction. The amount of
cytochrome c surface, i.e. Met-65, Met-80, His-18 and adduct formed is nearly the same. Also, there is some
His-33. evidence in both cases for the formation of additional
adducts where the [(triethylphosphine)gold(I)] moiety is
Aubipyc protein-bound.

When challenged with cytochrome c and lysozyme, Reactions of gold compounds with model proteins
Aubipyc manifested a quite constant behaviour that is in the presence of reducing agents
different from that of Auoxo6 and Au2phen. Small amounts
of metal–protein adducts were formed with both proteins; The results reported in the previous sections suggest that
in both cases the protein-bound molecular fragment was reduction of gold(III) may play a crucial role in the protein
the same, corresponding to the [(bipydmb-H)Au] moiety, metalation process, especially in the case of Auoxo6 and
i.e. the gold(III) centre plus the N,N,C terdentate ligand, as Au2phen. This observation led us to explore the effects of
proved by ESI MS analysis (Fig. 5). the addition of an excess of ‘‘physiological’’ reducing
That gold in the Aubipyc–protein adducts remains in the agents, such as ascorbic acid and GSH, to the various
?3 oxidation state is in good agreement with previous reaction batches on the assumption that a strongly reducing
results documenting the high redox stability of this or- environment might favour the protein metalation process.
ganogold(III) compound and its relatively scarce reactivity Indeed, it is well known that hypoxic cancer tissues exhibit
[25]. a frankly reducing microenviroment. Studies were restric-
ted to Auoxo6 and Au2phen because the gold(III) centre in
Auranofin Aubipyc is known to be highly resistant to reduction (see
earlier).
The reaction profiles of auranofin with the two model Auoxo6 and Au2phen were dissolved in the buffer in the
proteins were also investigated for comparative purposes. presence of either cytochrome c or lysozyme at standard
ESI MS spectra obtained at 72 h for auranofin–protein 3:1 molar ratios and ascorbic acid or GSH was added. The
adducts are shown in Fig. 6. The ESI MS spectra document concentration of the reducing agent was fixed at 1 mM as
well the formation of metallodrug–protein adducts, this value reflects the typical intracellular concentrations of
although in modest amounts. In both cases, the position of GSH. Remarkably, both reducing agents caused the char-
the major peak for the metal–protein adducts is consistent acteristic ligand-to-metal charge transfer bands of Auoxo6
with the intact auranofin molecule binding to protein, to disappear quickly, implying rapid reduction of the two

123
1300 J Biol Inorg Chem (2012) 17:1293–1302

Fig. 6 LTQ Orbitrap ESI mass spectra of auranofin dissolved in molar ratio of 3:1. Both spectra show peaks corresponding to the
25 mM TMeAmAc buffer, pH 7.4, in the presence of cyto- native protein (a) and to the formation of a [(triethylphos-
chrome c (A) or lysozyme (B) after 72 h incubation at 37 °C. The phine)gold(I)] moiety–protein adduct (Mr 315) (b) and an aurano-
protein concentration was 10-4 M (with a metal complex to protein fin–protein adduct with stoichiometries of 1:1 (c) and 2:1 (d)

gold(III) centres, and cytochrome c experienced simulta- established platinum(II) drugs, are scarcely reactive
neous conversion to its reduced form; important spectral towards DNA. Accordingly, their biological actions are
changes were also observed for Au2phen in line with those most probably the consequence of tight interactions
previously described for the reduction of Au2phen alone in occurring with a number of intracellular protein targets.
the reference buffer (Fig. S3). Inactivation of some crucial proteins might indeed consti-
The resulting metallodrug–protein samples (Auoxo6 and tute the ‘‘decisive event’’ ultimately triggering apoptotic
Au2phen reacted with cytochrome c or lysozyme) were cell death. In particular, the selenoenzyme thioredoxin
then analysed by ESI MS after short incubation times. reductase seems to be a good candidate protein target for
Comparative analysis of the spectra allows us to state that several cytotoxic gold compounds. A variety of other
addition of ascorbic acid invariantly causes the formation protein targets have been proposed [34–36], but conclusive
of far greater amounts of metal–protein adducts. At vari- target validation is still lacking.
ance, GSH, although accelerating the reduction of the In this work, we have analysed—systematically—the
gold(III) centres, apparently does not induce greater adduct interactions of three representative gold(III) compounds
formation. This may be explained by assuming that GSH at with two model proteins—i.e. lysozyme and cyto-
high concentrations (1 mM, as is the case here) is capable chrome c—to reveal the molecular details of the inherent
of sequestering most gold(I) ions in the form of Au(GSH)2 metalation processes. Comparative studies were conducted
complexes; yet, the presence of small quantities of adducts on auranofin, a clinically established antiarthritic gold(I)
in which gold(I) ions are attached to these two proteins drug. All these gold compounds typically behave as classic
could be demonstrated (Fig. S4). prodrugs; upon ‘‘chemical activation’’, they react with
proteins and form stable adducts. Complex activation may
simply consist of a ligand replacement reaction but also of
Discussion reductive disruption of the gold(III) centre and consequent
formation of ‘‘naked’’ gold(I) ions.
Gold compounds form a new class of promising cytotoxic The metalation processes of the model proteins men-
metallodrugs that are attracting growing attention within tioned were investigated—independently—by absorption
the scientific community as potential anticancer agents. spectroscopy and ESI MS methods. Absorption spectros-
The modes of action of cytotoxic gold compounds are still copy allowed us to monitor continuously the various
poorly understood. However, a number of observations metallodrug–protein systems over the whole incubation
suggest that cytotoxic gold compounds, at variance with time of 72 h. Conversely, ESI MS spectra recorded on

123
J Biol Inorg Chem (2012) 17:1293–1302 1301

metallodrug–protein samples at the end of the incubation processes that seem to play a central role in determining
period were particularly informative in revealing adduct the cellular effects of cytotoxic gold compounds (and more
formation and in determining the final metal-to-protein in general of various families of metal-based drugs). It is
stoichiometry and the nature of protein-bound metallic reasonable to assume that the cytotoxic gold(III) com-
fragments. plexes that were selected for the present investigation have
Protein metalation could be documented in most cases, the potential of metalating and altering a great number of
on the basis of the identification of a variety of metal– cellular proteins according to the mechanisms described
protein adducts in the ESI MS spectra. The number and herein; in turn, selective inactivation of a few crucial
the nature of the protein-bound metallic species were proteins (the true pharmacological targets) may ultimately
determined with accuracy. A rough estimate of the cause irreversible cell damage and death. A lot of experi-
amount of protein metalation was achieved by comparing mental effort is still needed to elucidate these issues in
the experimental peak intensities—i.e. the peak intensity depth, this being one of the major missions of modern
of the free protein versus the peak intensities of its metal metallomics.
adducts. The role of reducing agents in the protein metalation
Three distinct kinds of metallodrug–protein adducts processes was also assessed through a few additional
emerged from our studies. These adducts are formed experiments. Although it is evident that protein metalation
through one of the following mechanisms: (1) reduction of in two specific cases requires reduction of gold(III) to
gold(III), complex disruption and coordinative binding of gold(I), we established that the presence of an excess of
the resulting gold(I) ions to a typical protein donor, e.g. two distinct biological reducing agents, i.e. GSH and
histidine or methionine; (2) coordinative binding of a ascorbic acid, while causing rapid reduction of the gold(III)
redox-stable gold(III) fragment to protein donors; (3) non- centres, and also of cytochrome c, had contrasting effects
coordinative binding of the intact gold complex to the on the formation of metal–protein adducts. Ascorbic acid
protein. The pathway actually followed for adduct forma- greatly favoured adduct formation in the reactions of Au-
tion will typically depend on the nature of the metal oxo6 and Au2phen with cytochrome c or lysozyme; in
complex and the nature of the interacting protein. contrast, GSH did not exhibit a similar effect, possibly
Large differences were highlighted in the relative effi- because of extensive complexation of gold(I) by GSH
ciency of the various metalation processes and, accord- itself.
ingly, in the quantities of adducts formed. Differences in In conclusion, with the present study, we have tried to
protein metalation processes may be very relevant for the elucidate, at the molecular level, the processes of protein
effective mechanism of action of cytotoxic gold(III) com- metalation that occur when a few representative cytotoxic
pounds. Notably, the degree of protein metalation and the gold compounds react with two model proteins. A variety
resulting metal–protein stoichiometry critically depend on of situations were encountered depending on the nature of
both the nature of the protein and the nature of the metal the metal complex and the nature of the protein. From the
complex. The cases of the reactions of cytochrome c and results obtained, we can state that the process of adduct
lysozyme with Au2phen and Auoxo6 are particularly formation may follow a number of distinct routes leading
instructive; indeed, owing to the availability of a greater to a variety of structurally different adducts. However,
number of metal-binding side chains, cytochrome c gives prediction of which route will be preferred and what kinds
rise to adducts of higher metal-to-protein stoichiometry and amounts of metal–protein adducts will be formed for a
than lysozyme. Particularly interesting is the reaction of metallodrug–protein pair remains a very hard task; in fact,
Au2phen with cytochrome c that leads to the formation of the protein metalation processes appear to depend greatly
large amounts of a tetragold cytochrome c adduct where both on the nature of the metal complex and on the nature
four gold(I) ions are tightly associated with the protein. of the protein.
Further studies are under way to better characterise this It would be reasonable to envision that in defining the
adduct. in vivo pathways of metal-based drugs an important role is
Typically, our studies were conducted according to a played by proteins involved in the homeostasis of transition
standard protocol with a long incubation time of 72 h. metal ions such as copper, zinc and nickel and assembly of
However, some additional experiments were performed on their metal cofactors in metalloenzymes [37–40] but this
cytochrome c with shorter incubation times (see Figs. S5, aspect has been little characterised until now.
S6). Reduction of the incubation time resulted in a drastic We also showed that creating a strongly reducing
reduction in the amounts of adducts formed, with no evi- microenvironment resulted in variable effects on the pro-
dent change in the nature of the adducts. tein metalation processes depending on the nature of the
Overall, the present investigation stresses the biological reducing agent applied. It follows that each case of interest
and pharmacological relevance of protein metalation is yet to be investigated individually and that specific

123
1302 J Biol Inorg Chem (2012) 17:1293–1302

trends will hopefully be defined when a far larger mass of 20. Maiore L, Cinellu MA, Michelucci E, Moneti G, Nobili S,
experimental data are available. Landini I, Mini E, Guerri A, Gabbiani C, Messori L (2011) J
Inorg Biochem 105:348–355
21. Gabbiani C, Casini A, Kelter G, Cocco F, Cinellu MA, Fiebig
Acknowledgments We gratefully acknowledge Beneficentia Stif- HH, Messori L (2011) Metallomics 3:1318–1323
tung (Vaduz, Liechtenstein) and AIRC (IG-12085) for generous 22. Cinellu MA, Maiore L, Manassero M, Casini A, Arca M, Fiebig
financial support. HH, Kelter G, Michelucci E, Pieraccini G, Gabbiani C, Messori L
(2010) ACS Med Chem Lett 1:336–339
23. Gabbiani C, Casini A, Messori L, Guerri A, Cinellu MA, Min-
ghetti G, Corsini M, Rosani C, Zanello P, Arca M (2008) Inorg
References Chem 47:2368–2379
24. Cinellu MA, Minghetti G, Pinna MV, Stoccoro S, Zucca A,
Manassero M, Sansoni M (1998) J Chem Soc Dalton Trans
1. Nobili S, Mini E, Landini I, Gabbiani C, Casini A, Messori L
1735–1741
(2010) Med Res Rev 30:550–580
25. Marcon G, Carotti S, Coronnello M, Messori L, Mini E, Orioli P,
2. Gabbiani C, Casini A, Messori L (2007) Gold Bull 40:73–81
Mazzei T, Cinellu MA, Minghetti G (2002) J Med Chem
3. Tiekink ERT (2002) Crit Rev Oncol Hematol 42:225–248
45:1672–1677
4. Vergara E, Casini A, Sorrentino F, Zava O, Cerrada E, Rigobello
26. Casini A, Kelter G, Gabbiani C, Cinellu MA, Minghetti G,
MP, Bindoli A, Laguna M, Dyson PJ (2010) Chem Med Chem
Fregona DS, Fiebig HH, Messori L (2009) J Biol Inorg Chem
5:96–102
14:1139–1149
5. McKeage MJ, Maharaj L, Berners-Price SJ (2002) Coord Chem
27. http://www.oncotest.de/. Accessed 31 Oct 2012
Rev 232:127–135
28. Paull KD, Shoemaker RH, Hodes L, Monks A, Scudiero DA,
6. Casini A, Cinellu MA, Minghetti G, Gabbiani C, Coronnello M,
Rubinstein L, Plowman J, Boyd MR (1989) J Natl Cancer
Mini E, Messori L (2006) J Med Chem 49:5524–5531
81:1088–1092
7. Che C-M, Sun RW-Y, Yu WY, Ko CB, Zhu NY, Sun HZ (2003)
29. Huang RL, Wallqvist A, Covell DG (2005) Biochem Pharmacol
Chem Commun 1718–1719
69:1009–1039
8. Milacic V, Chen D, Ronconi L, Landis-Piwowar KR, Fregona D,
30. Zhang N, Du Y, Cui M, Xing J, Liu Z, Liu S (2012) Anal Chem
Dou QP (2006) Cancer Res 66:10478–10486
84(14):6206–6212
9. Gabbiani C, Messori L (2011) Anti Cancer Ageing Med Chem
31. Tanley SW, Schreurs AM, Kroon-Batenburg LM, Meredith J,
11:929–939
Prendergast R, Walsh D, Bryant P, Levy C, Helliwell JR (2012)
10. Mirabelli CK, Sung CM, Zimmerman JP, Hill DT, Mong S,
Acta Crystallogr D Biol Crystallogr 68(5):601–612
Crooke ST (1986) Biochem Pharmacol 35:1427–1433
32. Timerbaev AR, Pawlak K, Gabbiani C, Messori L (2011) Trends
11. Bindoli A, Rigobello MP, Scutari G, Gabbiani C, Casini A,
Anal Chem 30:1120–1138
Messori L (2009) Coord Chem Rev 253:1692–1707
33. Casini A, Guerri A, Gabbiani C, Messori L (2008) J Inorg Bio-
12. Rubbiani R, Kitanovic I, Alborzinia H, Can S, Kitanovic A,
chem 102:995–1006
Onambele LA, Stefanopoulou M, Geldmacher Y, Sheldrick WS,
34. Arnér ES, Holmgren A (2006) Semin Cancer Biol 6:420–426
Wolber G, Prokop A, Wölfl S, Ott I (2010) J Med Chem
35. Nguyen P, Awwad RT, Smart DD, Spitz DR, Gius D (2006)
53:8608–8618
Cancer Lett 236:164–174
13. Hickey JL, Ruhayel RA, Barnard PJ, Baker MV, Berners-Price
36. Vergara E, Casini A, Sorrentino F, Zava O, Cerrada E, Rigobello
SJ, Filipovska A (2008) J Am Chem Soc 130:12570–12571
MP, Bindoli A, Laguna M, Dyson PJ (2010) Chem Med Chem
14. Roberts JR, Xiao J, Schliesman B, Parsons DJ, Shaw CF III
5(1):96–102
(1996) Inorg Chem 35:424–433
37. Banci L, Bertini I, Cantini I, Ciofi-Baffoni S (2010) Cell Mol Life
15. Christodoulou J, Sadler PJ, Tucker A (1994) Eur J Biochem
Sci 67(15):2563–2589
225:363–368
38. Costello LC, Fenselau CC, Franklin RB (2011) J Inorg Biochem
16. Zou J, Taylor P, Dornan J, Robinson SP, Walkinshaw MD, Sadler
105(5):589–599
PJ (2000) Angew Chem Int Ed 39:2931
39. Zambelli B, Musiani F, Benini S, Ciurli S (2011) Acc Chem Res
17. Talib J, Beck JL, Ralph SF (2006) J Biol Inorg Chem 11:559–570
44(7):520–530
18. Marcon G, Messori L, Orioli P, Cinellu MA, Minghetti G (2003)
40. Zambelli B, Cremades N, Neyroz P, Turano P, Uversky V, Ciurli
Eur J Biochem 270:4655–4661
S (2012) Mol Biosyst 8:220–228
19. Messori L, Balerna A, Ascone I, Castellano C, Gabbiani C, Ca-
sini A, Marchioni C, Jaouen G, Congiu Castellano A (2011) J
Biol Inorg Chem 16:491–499

123

View publication stats

You might also like