SlideShare ist ein Scribd-Unternehmen logo
1 von 10
Downloaden Sie, um offline zu lesen
Full Paper
Evaluation of Novel Chalcone Oximes as Inhibitors of Tyrosinase
and Melanin Formation in B16 Cells
Sini K. Radhakrishnan, Ronald G. Shimmon, Costa Conn, and Anthony T. Baker
School of Chemistry and Forensic Science, University of Technology Sydney, Ultimo, NSW, Australia
A series of hydroxy-substituted chalcone oxime derivatives were synthesized. These compounds were
then evaluated for their inhibitory activities on tyrosinase and melanogenesis in murine B16F10
melanoma cells. The structures of the synthesized compounds were confirmed by 1
H NMR, 13
C NMR,
FTIR, and HRMS. Two of the compounds exhibited much higher tyrosinase inhibitory activities (IC50
values of 4.77 and 7.89 mM, respectively) than the positive control, kojic acid (IC50: 22.25 mM). Kinetic
studies revealed them to act as competitive tyrosinase inhibitors with their Ki values of 5.25 and
8.33 mM, respectively. Both the compounds inhibited melanin production and tyrosinase activity in
B16 cells. Docking results confirmed that the active inhibitors strongly interacted with the mushroom
tyrosinase residues.
Keywords: Chalcone oxime / Competitive / Docking / Tyrosinase inhibition
Received: August 24, 2015; Revised: October 14, 2015; Accepted: October 19, 2015
DOI 10.1002/ardp.201500298
:Additional supporting information may be found in the online version of this article at the publisher’s web-site.
Introduction
Melanogenesis is the process by which melanin, a polymeric
dark pigment, is produced and subsequently distributed by
melanocytes within the skin and hair follicles [1]. Melanin
plays a crucial role against skin photocarcinogenesis [2]. Excess
production of melanin from melanocytes can lead to
hyperpigmentation, which can manifest as melasma, cafe
au lait macules, ephelides (freckles), and dark spots [3–5]. The
development and screening of potent inhibitors of tyrosinase
is therefore of particular interest to the cosmetic industry.
Tyrosinase is the key enzyme involved in melanin biosyn-
thesis responsible for the hydroxylation of L-tyrosine to
L-DOPA (L‐3,4‐dihydroxyphenylalanine) and oxidation of
L-DOPA to DOPA quinone [6]. Previous reports confirmed
that tyrosinase was not only involved in melanizing in
animals, but also was one of the main causes for loss of
quality in fruits and vegetables during post-harvest handling
and processing, leading to faster degradation and shorter
shelf life [7, 8]. Additionally, tyrosinase may play a significant
role in neuromelanin formation in the human brain and could
be responsible for the neurodegeneration associated with
Parkinson’s disease [9]. Also, in insects, tyrosinase is uniquely
associated with three different biochemical processes, includ-
ing sclerotization of cuticle, defensive encapsulation and
melanization of foreign organisms, and wound healing [10].
Most melanin-biosynthesis inhibitors are phenol or catechol
analogs, which are structurally similar to the tyrosinase
substrates, tyrosine, or L-DOPA [11]. Besides, our interest
for chelator agents led us to study the hydroxamic acid group.
Hydroxamate molecules, one of the major classes of naturally
occurring metal complexing agents, have been thoroughly
studied as ligands for different metal ions such as Fe(III), Zn(II),
and Cu(II) [12, 13]. The chelation involves the oxygen
belonging to the ––N–OH group [14]. Previously, we have
reported the tyrosinase inhibitory potential of novel aza-
chalcone compounds [15]. The presence in chalcones of a
conjugated double bond and a completely delocalized p
electron system reduces their redox potentials and makes
them susceptible to electron transfer reactions. The a–b
unsaturated bond enables the chalcone to act as a Michael
acceptor for nucleophilic species including glutathione (GSH)
Correspondence: Dr. Sini K. Radhakrishnan, School of Chemistry
and Forensic Science, University of Technology Sydney, 15
Broadway, Ultimo, NSW 2007, Australia.
E-mail: Sini.KaranayilRadhakrishnan@student.uts.edu.au
Fax: þ61295142000
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 1
or cysteine residues on proteins [16]. Previous studies have
indicated the use of chalcone oximes as anticancer agents [17].
This prompted us to further investigate the effect of chalcone
oximes on melanin formation in murine B16F10 cells.
From a structural perspective, tyrosinase has two copper
ions in its active site which play a vital role in its catalytic
activity. Each of the copper ions is coordinated by three
histidines in the protein matrix [18]. At the active site of
tyrosinase, a dioxygen molecule binds in a side-on coordina-
tion mode between two copper ions. The copper atoms
participate directly in hydroxylation of monophenols to
diphenols (cresolase activity) and in the oxidation of
o-diphenols to o-quinones (catechol oxidase activity) that
enhance the production of the brown color [19, 20]. Inhibition
of the enzyme tyrosinase has been a major motivation for
researchers, and several inhibitors are currently used as
cosmetic additives and medicinal products for the treatment
of hyperpigmentation. Although hydroquinone has been the
mainstay treatment for hyperpigmentation, its clinical poten-
tial has been complicated by a number of adverse reactions
including contact dermatitis, irritation, transient erythema,
burning, prickling sensation, leukoderma, chestnut spots on
the nails, hypochromia, and ochronosis [21–25]. Moreover,
hydroquinone is potentially mutagenic to mammalian cells.
Hydroquinone and its derivative arbutin are both catabolized
to benzene metabolites with the potential for bone marrow
toxicity [26]. Another alternative, kojic acid is currently
applied as a cosmetic skin whitener and food additive to
prevent enzymatic browning [27]. However, its use in
cosmetics has been limited, because of its instability during
storage [28]. Further investigation of potential tyrosinase
inhibitors that could be applied during food processing to
curb enzymatic browning and for the treatment of hyperpig-
mentation disorders is warranted.
Results and discussion
Chemistry and inhibition kinetics
In the current study, we report the successful identification of
several hydroxy-substituted chalcone oxime compounds with
improved tyrosinase inhibition. A series of hydroxy-substi-
tuted chalcone compounds were synthesized by base cata-
lyzed Claisen–Schmidt condensation of an aldehyde and an
appropriate ketone in a polar solvent like methanol. These
compounds were then subsequently converted to their
oxime derivatives by reaction with hydroxylamine hydrochlo-
ride (Scheme 1). The structures of the compounds synthesized
were confirmed by 1
H NMR, 13
C NMR, FTIR, and HRMS. Assays
were performed with L-DOPA as the substrate, using kojic
acid, a well-known effective tyrosinase inhibitor as the
positive control. The inhibitory activities of the synthesized
oxime compounds 2a–g were compared with the reference
compound kojic acid (Fig. 1).
In terms of the structure–activity relationships, compound
2b (2-[(1E,2E)-N-hydroxy-3-(2-methoxy-4-nitrophenyl)prop-2-
enimidoyl]phenol) exhibited the most potent tyrosinase
inhibitory activity with inhibition of 80.19%. This could be
accounted to the presence of a strongly activating ortho-
methoxy substituent and a meta directing nitro substituent.
The presence of a strong electron releasing group in the ortho
position (–OMe) and an electron withdrawing group (–NO2) in
the para position modulates the electronic structure of ring B
significantly. It is equally likely for the oxime moiety to
coordinate with the copper atoms present in the tyrosinase
active site. Also, shifting the methoxy substituent to the para
position in ring B led to a decrease in tyrosinase inhibitory
activity as exhibited by compound 2d. Replacement of the
phenyl ring with a pyridine ring brought about a decline in
tyrosinase inhibition (60.6%) as seen with compound 2a. This
shows that ring A is not oriented such that the pyridine
nitrogen can bind the copper center when compared to kojic
acid. Compound 2e with an electron donating para-dimethyl
amino group on ring B showed an inhibition of 72.12% at
20 mM. The inhibition of both compounds 2b and 2e was seen
to be greater than the positive control kojic acid which
exhibited 47.67% inhibition at 20 mM. Compounds 2a, 2c, and
2d showed significant tyrosinase inhibitions. Replacement of
phenyl ring with a naphthyl ring gave compound 2f with
moderate tyrosinase inhibition whereas compound 2g
showed low inhibitory activity (50.6%) against mushroom
tyrosinase. In particular, compounds 2b and 2e exhibited the
greatest inhibition of L-DOPA oxidase activity of mushroom
tyrosinase.
In this study, we investigated in depth the tyrosinase
inhibitory activities of compounds 2b and 2e. As shown in
Table 1, compounds 2b, 2e, and kojic acid inhibited mushroom
tyrosinase activity in a concentration-dependent manner. Both
the compounds 2b and 2e had better inhibitory activities with
IC50 values of 4.77 and 7.89mM, respectively, than
the reference compound kojic acid (IC50: 22.25 mM). To explore
the mechanism, we examined the kinetics of tyrosinase
inhibition in the presence of the most active compounds 2b
and 2e. The data are shown in Table 2. As the concentrations of
active inhibitors 2b and 2e increased, Km values gradually
increased, but Vmax values did not change, thereby indicating
that the inhibitors act as competitive inhibitors of mushroom
tyrosinase (Fig. 2). Dixon plots gave a family of straight lines
passing through the same point at the second quadrant, giving
the inhibition constant (Ki) (Fig. 3). The Ki value estimated from
this Dixon plot was 5.25 and 8.33mM for the compounds 2b and
2e, respectively. Kinetic results indicate that compounds 2b and
2e might inhibit the tyrosinase activity by binding to the copper
containing active site region. To confirm this, we have further
simulated docking of these active compounds with mushroom
tyrosinase.
Docking results
The Accelrys Discovery Studio 4.5 suite was utilized to simulate
binding between mushroom tyrosinase and active tyrosinase
inhibitors 2band2e. Tomodelthetyrosinasestructure,weused
the crystal structure of Agaricus bisporus (mushroom)
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
S. K. Radhakrishnan et al. Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 2
tyrosinase (PDB ID: 2Y9X) A chain. Among all the compounds
synthesized, compounds 2b and 2e had the highest docking
scoreswhencomparedwiththestandard,kojicacid(dockscore:
À10.29 kcal/mol). Docking results showed that compound 2b
hadbetterbindingcapability(dockscore:À28.94 kcal/mol)with
mushroomtyrosinaseascomparedtocompound2e(dockscore:
À27.45 kcal/mol) (Fig. 4). The greater inhibitory efficacy of
compound 2b is attributed to chelation of the oxime oxygen
atomwiththecopperion(Cu401)intheactivesiteofmushroom
tyrosinase (2.29 Å). We searched for tyrosinase residues that
might bind to compounds 2b and 2e, and found that the most
important binding residues for interaction are His65, His85,
Val248, Ala286, His244, Asn260, Val283, Met280, Ser282,
His259, Phe292, and Glu322 for both compounds 2b and 2e.
Strong hydrogen bonding with His61, His85, and Val283 was
seen for compound 2b whereas compound 2e exhibited
hydrogen bonding interactions with Asn260, Glu322, and
His259. Both the active compounds showed hydrophobic p–p
stacking interactions with His263 and T-shaped edge to face
aryl–aryl interactions with Phe264 and His244.
In vitro cytotoxic studies
Our work was further extended to investigate the effect of
novel chalcone oxime compounds 2b and 2e on the
tyrosinase activity and melanin levels in B16F10 melanoma
cells. The cytotoxic effects of 2b and 2e were estimated by
measuring cell viability, where no significant cytotoxic effect
Scheme 1. General method for the synthesis of hydroxy-substituted chalcones and their oxime derivatives. Reagents and conditions:
(a) MeOH, NaOH, 0°C, 24 h; (b) NH2OH.HCl.
0
10
20
30
40
50
60
70
80
90
2g2f2e2d2c2b2aKA
TyrosinaseinhibiƟon(%,20μM)
Compound
***
***
***
*** *** ***
*
Figure 1. Inhibitory effects of chalcone oximes on mushroom
tyrosinase activity. Values represent means Æ SE of three
determinations. Inhibitory concentration is 20 mM. Ã
p  0.05
and ÃÃÃ
p  0.001 compared to kojic acid. KA, kojic acid.
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 3
was found at any of the concentrations tested. At doses of
1.0, 10.0, 50.0, and 100.0 mM of compounds 2b or 2e for 48 h,
cell viability was 118, 92.2, 78.4, and 72.6% for compound 2b
and 89, 82, 76.4, and 64% for compound 2e, respectively,
compared with the control (Fig. 5). Thus, neither compound
2b nor 2e were cytotoxic to B16 cells in the concentration
range of 1.0–100.0 mM. We then ascertained the inhibitory
effects of compounds 2b and 2e on melanogenesis by
quantifying the melanin content of B16 cells treated with
compounds 2b and 2e (Fig. 6). The melanin content of B16
cells after treatment with compound 2b in the presence of
100 nM a-melanocyte-stimulating hormone (a-MSH) de-
creased dose-dependently, showing 215.25% at 1.0 mM,
175.16% at 5.0 mM, and 112.0% at 10.0 mM, compared with
the 100 nM a-MSH-only treated group (225.22%) and the
control group (100%). Similarly, cells treated with compound
2e exhibited melanin contents of 235.17% at 1.0 mM,
215.55% at 5.0 mM, and 165.35% at 10.0 mM compared with
100 nM a-MSH-only-treated group (225.22%) and the
control group (100%).
Finally, we determined the inhibitory effects of compounds
2b and 2e on tyrosinase activity of B16 cells treated with
100 nm a-MSH. After 24 h incubation with 2b and 2e,
tyrosinase activities were 145.91% at 1.0 mM, 114.52% at
5.0 mM, and 108.77% at 10.0 mM of 2b (Fig. 7), compared with
the 100 nM a-MSH-only treated group (225.25%) and the
control group (100%). Similarly, cells treated with compound
2e exhibited melanin contents of 160.45% at 1.0 mM, 126.5%
at 5.0 mM, and 111.22% at 10.0 mM compared with 100 nM
a-MSH-only-treated group (225.25%) and the control group
(100%). The inhibition of compounds 2b and 2e on murine
tyrosinase activity was greater than the reference compound
kojic acid. The results establish that compounds 2b and 2e
suppress melanin biosynthesis through the inhibition of
tyrosinase.
Conclusion
To summarize, among hydroxy-substituted chalcone oxime
compounds, compounds 2b and 2e were found to be more
potent than the positive reference compound kojic acid.
Kinetic studies identified both the compounds to be
competitive inhibitors of mushroom tyrosinase. Docking
simulation studies determined the possible residues of
tyrosinase that could play a key role in binding the inhibitor
molecules. In the B16 cell culture system, tyrosinase activity
and melanin levels were significantly reduced in the presence
of compounds 2b and 2e, suggesting therefore inhibition of
melanin biosynthesis to be attributed to tyrosinase inhibition.
The structures of the non-cyclic moieties of our molecules
have similar features to those of hydroxamic acids and
hydroxamates which are good chelating agents. Tyrosinase
inhibition of compounds 2b and 2e depended on the
competency of the inhibitor compounds to block access by
any substrate molecule in the active site of the enzyme. Strong
coordination with the copper metal, significant hydrophobic
and p–p stacking interactions contributed to effective
Table 2. Kinetic analysis of compounds 2b and 2e.
Compound Type of inhibition Vmax (mM/min) Ki (mM)a)
2b Competitive 0.045 5.25
2e Competitive 0.023 8.33
a)
Data are represented as mean values of three independent tests. Ki is the (inhibitor constant). Vmax is the maximum reaction
velocity.
Table 1. Inhibitory effects of kojic acid, 2b, and 2e on mushroom tyrosinase activity.
Sample
Concentration
(mM) Inhibition (%)
Average of
inhibition (%) IC50 (mM)a)
Kojic acid 1.25 2.19 3.90 2.65 2.91 22.25 Æ 0.22
5.00 29.22 28.53 30.22 29.32
20.00 49.26 47.54 46.23 47.67
2b 1.25 43.20 44.17 42.65 43.34 4.77 Æ 0.35
5.00 49.60 50.25 57.44 52.43
20.00 80.60 78.77 81.22 80.19
2e 1.25 39.36 34.25 35.50 36.37 7.89 Æ 0.15
5.00 45.22 47.21 44.65 45.69
20.00 72.50 70.23 73.65 72.12
a)
Fifty percent inhibitory concentration (IC50).
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
S. K. Radhakrishnan et al. Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 4
tyrosinase inhibition. Also, oximes, popular in supramolecular
chemistry as strong hydrogen bond donors, were involved in
hydrogen bond interactions with the amino acid residues of
the enzyme [29]. This, in turn, helped the compound to have a
better fit to the catalytic pocket of the enzyme tyrosinase. The
current study portrays chalcone oximes as promising candi-
dates to be explored as depigmentation agents.
Experimental
Chemistry
Chemical reagents and instruments
Melting points (mp) were determined with WRS-1B melting
point apparatus and the thermometer was uncorrected. NMR
spectra were recorded on Agilent 500 spectrometer at 25°C in
CDCl3 or DMSO-d6. All chemical shifts (d) are quoted in parts
per million downfield from TMS and coupling constants (J) are
given in Hertz. Abbreviations used in the splitting pattern
were as follows: s ¼ singlet, d ¼ doublet, t ¼ triplet, m ¼ mul-
tiplet. HRMS spectra were recorded using the Agilent
Technologies 6520 LC/MS-QTOF. All reactions were monitored
by TLC (Merck Kieselgel 60 F254) and the spots were visualized
under UV light. Infrared (IR) spectra were recorded on Thermo
Scientific NICOLET 6700 FT-IR spectrometer. Tyrosinase, L-3,4-
dihydroxyphenylalanine (L-DOPA) and kojic acid were pur-
chased from Sigma–Aldrich Chemical Co.
General method for the synthesis of hydroxy chalcone
derivatives (1a–g)
To a stirred solution of the appropriate ketone (1 mM) and a
substituted aldehyde (1 mM) in 25 mL methanol, was added
pulverized NaOH (2 mM) and the mixture was stirred at room
temperature for 24–36 h. The reaction was monitored by TLC
using n-hexane/ethyl acetate (7:3) as mobile phase. The
reaction mixture was cooled to 0°C (ice-water bath) and
acidified with HCl (10% v/v aqueous solution) to afford total
precipitation of the compounds. In most cases, a yellow
precipitate was formed, which was filtered and washed with
10% aqueous HCl solution. In the cases where an orange oil
was formed, the mixture was extracted with CH2Cl2, the
extracts were dried (Na2SO4), and the solvent was evaporated
to give the respective chalcone (1a–g).
(2E)-1-(2-Hydroxyphenyl)-3-(2-methoxy-4-nitrophenyl)-
prop-2-en-1-one 1a
Mp: 123–125°C; 1
H NMR (500 MHz, CDCl3): d 12.22 (s, 1H, OH),
8.22 (s, 1H), 7.95 (d, 1H, J ¼ 14.0, Hb,), 7.86 (m, 1H, J ¼ 9.5, H-6),
7.80 (d, 1H, J ¼ 14.0, Ha), 7.75 (d, 1H, J ¼ 10.5, H-60
), 7.69 (d, 1H,
J ¼ 8.5, H-5), 7.59 (dd, 1H, J ¼ 8.5, H-40
), 7.02 (dd, 1H, J ¼ 9.0, H-
-200
-100
0
100
200
300
400
-4 -3 -2 -1 0 1 2 3 4 5 6
1/V(µM/min)-1
1/S (mM/L)
20 µM
5 µM
1.25 µM
No inhibitor
2b
-200
-100
0
100
200
300
400
-4 -2 0 2 4 6
1/V(mM/min)-1
1/S (mM/L)
20 µM
5 µM
1.25 µM
No inhibitor
2e
Figure 2. Lineweaver–Burk plot for inhibi-
tion of compounds 2b and 2e on mushroom
tyrosinase. Data were obtained as mean
values of 1/V, the inverse of the absorbance
increase at a wavelength of 492 nm/min of
three independent tests with different con-
centrations of L-DOPA as a substrate. The
concentrations of compounds 2b and 2e from
top to bottom are 20, 5, 1.25, and 0 mM,
respectively.
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 5
30
), 6.97 (m, 1H, J ¼ 10.0, H-50
), 3.82 (s, 3H); 13
C NMR (125 MHz,
DMSO-d6) d 194.2 (C––O), 130.2 (C60
), 119.33 (C10
), 118.2 (C50
),
137.0 (C40
), 118.4 (C30
), 162.9 (C20
), 128.2 (C1), 115.2 (C2), 112.2
(C3), 137.5 (C4), 122.4 (C5), 116.5 (C6), 125.5 (vinylic), 140.8
(vinylic); IR (KBr) n (cmÀ1
): 3250, 3345, 3015, 2910, 2865, 2700,
1702, 1682, 1552, 1465, 1305, 979, 735, 680, 575; MS (ESI):
270.1 ([MþH])þ
.
(2E)-3-(2-Methoxy-4-nitrophenyl)-1-(pyridin-2-yl)prop-2-
en-1-one 1b
Mp: 126–128°C; 1
H NMR (500 MHz, CDCl3): d 8.70 (dd, 1H,
J ¼ 10.0, H-3), 8.32 (dd, 1H, J ¼ 8.5, H-6), 7.90 (d, 1H, J ¼ 12.0,
Ha), 8.17 (s, 1H), 7.94 (m, 1H, J ¼ 8.0, H-60
), 7.87 (t, 1H, J ¼ 9.0,
H-5), 7.79 (d, 1H, J ¼ 10.5, H-50
), 7.40 (m, 1H, J ¼ 10.5, H-4),
6.95 (d, 1H, J ¼ 12.0, Hb), 3.86 (s, 3H); 13
C NMR (125 MHz,
DMSO-d6) d 195.5 (C––O), 155.2 (C1), 120.9 (C6), 136.2 (C5),
129.4 (C4), 150.2 (C3), 134.0 (C10
), 116.2 (C20
), 112.2 (C30
),
138.2 (C40
), 125.3 (C50
), 118.2 (C60
), 140.2 (vinylic), 129.2
(vinylic); IR (KBr) n (cmÀ1
): 3015, 2952, 1669, 1640, 1405, 1355,
1265, 1175, 805, 672; MS (ESI): 285.1 ([MþH])þ
.
(2E)-1-(2-Hydroxyphenyl)-3-(4-methoxy-3-nitrophenyl)-
prop-2-en-1-one 1c
Mp: 110–112°C; 1
H NMR (500 MHz, CDCl3): d 12.47 (s, 1H, OH),
8.10 (s, 1H), 7.86 (d, 1H, J ¼ 12.5, Hb), 7.74 (d, 1H, J ¼ 10.0, H-60
),
7.72 (d, 1H, J ¼ 12.5, Ha), 7.65 (d, 1H, J ¼ 9.5, H-5), 7.54 (d, 1H,
J ¼ 8.0, H-6), 7.49 (t, 1H, J ¼ 8.5, H-40
), 6.92 (dd, 1H, J ¼ 9.5, H-
30
), 6.85 (dd, 1H, J ¼ 9.0, H-50
), 3.86 (s, 3H); 13
C NMR (125 MHz,
DMSO-d6) d 192.2 (C––O), 130.5 (C60
), 119.2 (C10
), 118.2 (C50
),
136.7 (C40
), 118.2 (C30
), 161.9 (C20
), 126.9 (C1), 120.5 (C2), 137.2
(C3), 147.5 (C4), 112.4 (C5), 114.2 (C6), 56.6 (Me), 122.4
(vinylic), 141.8 (vinylic); IR (KBr) n (cmÀ1
): 3200, 3070, 2914,
2864, 2720, 1686, 1578, 1550, 1468, 1349, 970, 720, 580; MS
(ESI): 270.1 ([MþH])þ
.
(2E)-3-(4-Methoxy-3-nitrophenyl)-1-(pyridin-2-yl)prop-2-
en-1-one 1d
Mp: 116–118°C; 1
H NMR (500 MHz, CDCl3): d 8.75 (dd, 1H,
J ¼ 9.5, H-30
), 8.25 (dd, 1H, J ¼ 8.5, H-60
), 8.17 (s, 1H), 8.12 (s,
-40
-30
-20
-10
0
10
20
30
40
-40 -30 -20 -10 0 10 20 30
1/v(µM/min)-1
[inhibitor] µM
2e
-40
-30
-20
-10
0
10
20
30
40
50
-40 -30 -20 -10 0 10 20 30
1/v(µM/min)-1
[inhibitor] µM
2b
Figure 3. Dixon plot for the inhibitory effect of compounds 2b
and 2e on L-DOPA oxidation catalyzed by mushroom tyrosinase.
The inhibitor concentrations were 0, 10, and 20 mM, res-
pectively. The L-DOPA concentrations were 200, 400, and
600 mM.
Figure 4. Docking results of compounds 2b and 2e in the
tyrosinase catalytic pocket. Ligands 2b and 2e are displayed as
ball and stick whereas the core amino acid residues are
displayed as stick. The green-dotted lines show the hydrogen
bond interactions and the purple lines show the non-bonding
interactions. The ochre balls represent the copper ions.
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
S. K. Radhakrishnan et al. Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 6
1H), 7.89 (d, 1H, J ¼ 13.5, Ha), 7.86 (t, 1H, J ¼ 9.5, H-50
), 7.69 (d,
1H, J ¼ 9.0, H-5), 7.57 (d, 1H, J ¼ 8.5, H-6), 7.47 (dd, 1H, J ¼ 11.0,
H-40
), 6.94 (d, 1H, J ¼ 13.5, Hb), 3.90 (s, 3H); 13
C NMR (125 MHz,
DMSO-d6) d 162.2 (C––N), 154.4 (C1), 121.2 (C6), 134.2 (C5),
130.4 (C4), 148.2 (C3), 135.4 (C10
), 118.2 (C20
), 114.2 (C30
), 140.7
(C40
), 125.3 (C50
), 120.4 (C60
), 139.2 (vinylic), 129.7 (vinylic); IR
(KBr) n (cmÀ1
): 3020, 2955, 1670, 1640, 1435, 1350, 1252, 1175,
805, 672; MS (ESI): 285.1 ([MþH])þ
.
(2E)-3-[4-(Dimethyl amino)phenyl]-1-(2-hydroxyphenyl)-
prop-2-en-1-one 1e
M.p: 108–110°C; 1
H NMR (500 MHz CDCl3): d 12.26 (s, 1H, OH),
8.06 (d, 1H, J ¼ 11.5, Hb), 7.85 (d, 1H, J ¼ 11.5, Ha), 7.72 (d, 1H,
J ¼ 5.5, H-6), 7.68 (dd, 2H, J ¼ 8.0, H-20
and H-60
), 7.49 (dd, 1H,
J ¼ 8.5, H-4), 6.92 (t, 1H, J ¼ 9.0, H-3), 6.89 (dd, 1H, J ¼ 15.0, H-
5), 6.64 (dd, 2H, J ¼ 9.0, H-30
and H-50
); 13
C NMR (100 MHz,
DMSO-d6) d 190.3 (C––O), 125.1 (C1), 131.9 (C2 and C6), 110.9
(C5 and C3), 154.3 (C4), 40.0 (N–CH3), 145.3 (vinylic), 125.1
(vinylic), 119.7 (C10
), 162.3 (C20
), 118.1 (C30
), 118.3 (C40
), 136.4
(C50
), 130.9 (C60
); IR (KBr) n (cmÀ1
): 3420, 1710, 1640, 1335, 813,
680, 757, 680; MS (ESI): 268.2 ([MþH])þ
.
(2E)-1-(3-Hydroxynaphthalen-2-yl)-3-(2-methoxy-4-
nitrophenyl)prop-2-en-1-one 1f
Mp: 115–117°C; 1
H NMR (500 MHz, CDCl3): d 12.69 (s, 1H, OH),
8.37 (dd, 1H, J ¼ 10.5, H-50
), 8.20 (dd, 1H, J ¼ 9.0, H-100
), 7.68
(dd, 1H, J ¼ 9.5, H-80
), 7.60 (dd, 1H, J ¼ 8.5, H-60
), 7.57 (d, 1H,
J ¼ 12.0, Hb), 7.52 (d, 1H, J ¼ 12.0, Ha), 7.40 (m, 1H, J ¼ 10.0, H-
70
), 7.25 (m, 1H, J ¼ 8.5, H-30
); 13
C NMR (125 MHz, DMSO-d6) d
194.5 (C––O), 125.4 (C10
), 159.8 (C20
), 119.2 (C30
), 130.2 (C40
),
125.4 (C50
), 132.5 (C60
), 126.4 (C70
), 132.4 (C80
), 130.2 (C90
),
144.2 (C100
), 130.2 (vinylic), 149.8 (vinylic), 55.6 (Me), 130.2
(C1), 119.8 (C2), 117.4 (C3), 130.4 (C4), 124.7 (C5), 119.5 (C6); IR
(KBr) n (cmÀ1
): 3350, 3249, 3017, 2850, 2650, 1685, 1605, 1552,
1450, 1325, 975, 728, 687, 568; HRMS m/z: 350.1009 ([MþH])þ
;
calcd.: 350.1022.
(2E)-1-(3-Hydroxynaphthalen-2-yl)-3-(4-methoxy-3-
nitrophenyl)prop-2-en-1-one 1g
Mp: 112–114°C; 1
H NMR (500 MHz, CDCl3): d 12.29 (s, 1H, OH),
8.12 (dd, 1H, J ¼ 8.0, H-100
), 8.15 (s, 1H), 8.07 (dd, 1H, J ¼ 10.5,
H-50
), 7.90 (dd, 1H, J ¼ 10.5, H-80
), 7.79 (d, 1H, J ¼ 13.5, Hb), 7.65
(d, 1H, J ¼ 9.0, H-5), 7.62 (d, 1H, J ¼ 13.5, Ha), 7.50 (dd, 1H,
J ¼ 9.0, H-60
), 7.42 (d, 1H, J ¼ 8.5, H-6), 7.39 (m, 1H, J ¼ 7.5, H-
70
), 7.24 (dd, 1H, J ¼ 9.5, H-30
), 3.72 (s, 3H); 13
C NMR (125 MHz,
DMSO-d6) d 195.4 (C––O), 120.8 (C10
), 162.4 (C20
), 119.4 (C30
),
130.2 (C40
), 125.8 (C50
), 130.1 (C60
), 124.4 (C70
), 130.4 (C80
),
130.2 (C90
), 142.25 (C100
), 128.9 (vinylic), 147.5 (vinylic), 127.5
0
20
40
60
80
100
120
140
1 μM 10 μM 50 μM 100 μM 1 μM 10 μM 50 μM 100 μM control
e2b2
)lortnocfo%(ytilibaivlleC
Figure 5. Effect of compounds 2b and 2e on cell viability. Data
are expressed as a percentage of the control.
0
50
100
150
200
250
300
control
α-MSH
α-MSH+1.0μM
α-MSH+5.0μM
α-MSH+10.0μM
control
α-MSH
α-MSH+1.0μM
α-MSH+5.0μM
α-MSH+10.0μM
α-MSH+kojicacid10.0μM
2e2b
melanincontent(%ofcontrol)
### ***
***
***
***
*** ***
***###
Figure 6. Inhibitory effect of compounds 2b and 2e after
treatment with 100 nM a-MSH in B16 cells. Melanin contents
were measured at 405 nm. Values represent the mean Æ SE of
three experiments. Data are expressed as a percentage of the
control. ÃÃÃ
p  0.001 compared to the group treated with
100 nm a-MSH and ###
p  0.001, compared with the untreated
control.
0
50
100
150
200
250
300
control
α-MSH
α-MSH+1.0μM
α-MSH+5.0μM
α-MSH+10.0μM
control
α-MSH
α-MSH+1.0μM
α-MSH+5.0μM
α-MSH+10.0μM
α-MSH+kojicacid10.0μM
e2b2
Tyrosinaseactivity(%ofcontrol)
***
***
***
*** *** ***
***
### ###
Figure 7. Inhibitory effect of compounds 2b and 2e on B16 cells
tyrosinase. Values represent the mean Æ SE of three experi-
ments. Data are expressed as a percentage of the control.
ÃÃÃ
p  0.001 compared to the group treated with 100 nm a-MSH
and ###
p  0.001, compared with the untreated control.
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 7
(C1), 122.5 (C2), 139.2 (C3), 147.4 (C4), 114.2 (C5), 114.2 (C6),
59.3 (Me); IR (KBr) n (cmÀ1
): 3255, 3065, 2924, 2847, 2720,
2650, 1672, 1570, 1550, 1425, 1315, 968, 725, 580; HRMS m/z:
350.1012 ([MþH])þ
; calcd.: 350.1022.
General method for the synthesis of hydroxy chalcone
oxime compounds (2a–g)
A mixture of chalcones 1a–g (1 mmol), hydroxylamine
hydrochloride (1.5 mmol), and anhydrous sodium sulfate
(1 mmol) was refluxed in ethanol (5 mL) under stirring for
the period (the reaction was followed by TLC). After
completion of the reaction, the mixture was filtered and
the solvent was evaporated under reduced pressure. Then
distilled water (10 mL) was added, and ethyl acetate (10 mL
 3) was added to extract organic compounds. The combined
organic layers were dried over anhydrous sodium sulfate and
filtered. Evaporation of the solvent in reduced pressure gave
the crude product, which was purified by column chromatog-
raphy on silica (200–300 mesh), eluted with petroleum ether
(60–90°C), or a mixture of petroleum ether and ethyl acetate
(v/v ¼ 1:4) to yield the pure products (2a–g).
2-[(1E,2E)-N-Hydroxy-3-(2-methoxy-4-nitrophenyl)prop-2-
enimidoyl]phenol 2a
Mp: 190À192°C; 1
H NMR (500 MHz, CDCl3): d 12.52 (s, 1H, OH),
9.47 (s, 1H), 8.05 (s, 1H), 7.90 (m, 1H, J ¼ 8.5, H-6), 7.87 (d, 1H,
J ¼ 12.5, Ha), 7.77 (d, 1H, J ¼ 11.0, H-60
), 7.72 (d, 1H, J ¼ 9.5, H-
5), 7.50 (dd, 1H, J ¼ 8.0, H-40
), 7.24 (dd, 1H, J ¼ 9.5, H-30
), 6.94
(dd, 1H, J ¼ 10.5, H-50
), 6.89 (d, 1H, J ¼ 12.5, Hb), 3.89 (s, 3H);
13
C NMR (125 MHz, DMSO-d6) d 160.2 (C––N), 130.44 (C60
),
120.43 (C10
), 118.3 (C50
), 139.2 (C40
), 118.4 (C30
), 164.9 (C20
),
132.2 (C1), 114.9 (C2), 112.2 (C3), 137.5 (C4), 124.7 (C5), 118.5
(C6), 129.4 (vinylic), 142.5 (vinylic); IR (KBr) n (cmÀ1
): 3650,
3582, 2650, 1648, 1550, 1265, 949, 835, 712; HRMS m/z:
315.0976 ([MþH])þ
; calcd.: 315.0981.
(1E,2E)-N-Hydroxy-3-(2-methoxy-4-nitrophenyl)-1-
(pyridin-2-yl)prop-2-en-1-imine 2b
Mp: 178–180°C; 1
H NMR (500 MHz, CDCl3): d 9.42 (s, 1H), 8.77
(dd, 1H, H-3, J ¼ 10.5), 8.47 (dd, 1H, H-6, J ¼ 8.0), 8.20 (s, 1H),
7.88 (m, 1H, J ¼ 8.0, H-60
), 7.85 (d, 1H, J ¼ 11.5, Ha), 7.80 (t,1H,
H-5, J ¼ 9.5), 7.75 (d, 1H, J ¼ 9.5, H-50
), 7.44 (dd, 1H, J ¼ 11.0, H-
4), 6.94 (d, 1H, J ¼ 11.5, Hb), 3.88 (s, 3H); 13
C NMR (125 MHz,
DMSO-d6) d 162.5 (C––N), 153.5 (C1), 118.9 (C6), 134.4 (C5),
130.4 (C4), 150.2 (C3), 136.4 (C10
), 118.2 (C20
), 114.2 (C30
), 139.7
(C40
), 125.3 (C50
), 120.2 (C60
), 144.2 (vinylic), 133.2 (vinylic); IR
(KBr) n (cmÀ1
): 3645, 3548, 2652, 1635, 1425, 1302, 1225, 805,
672; HRMS m/z: 300.0978 ([MþH])þ
; calcd.: 300.0984.
2-[(1E,2E)-N-Hydroxy-3-(4-methoxy-3-nitrophenyl)prop-2-
enimidoyl]phenol 2c
Mp: 165–167°C; 1
H NMR (500 MHz, CDCl3): d 12.50 (s, 1H, OH),
9.39 (s, 1H), 8.22 (s, 1H), 7.92 (d, 1H, J ¼ 11.5, Ha), 7.82 (d, 1H,
J ¼ 10.0, H-60
), 7.64 (d, 1H, J ¼ 9.0, H-5), 7.57 (d, 1H, J ¼ 8.0, H-
6), 7.44 (t, 1H, J ¼ 8.0, H-40
), 7.35 (d, 1H, Hb, J ¼ 11.5), 7.02 (dd,
1H, J ¼ 9.5, H-50
), 6.94 (dd, 1H, J ¼ 10.5, H-30
), 3.92 (s, 3H);
13
C NMR (125 MHz, DMSO-d6) d 160.2 (C––N), 129.9 (C60
), 120.2
(C10
), 118.1 (C50
), 137.7 (C40
), 118.2 (C30
), 162.9 (C20
), 124.9 (C1),
118.4 (C2), 137.2 (C3), 148.5 (C4), 114.2 (C5), 116.3 (C6), 55.8
(Me), 123.2 (vinylic), 140.9 (vinylic); IR (KBr) n (cmÀ1
): 3659,
3545, 2650, 1625, 1400, 1312, 1234, 875, 632; HRMS m/z:
315.0975 ([MþH])þ
; calcd.: 315.0981.
(1E,2E)-N-Hydroxy-3-(4-methoxy-3-nitrophenyl)-1-
(pyridin-2-yl)prop-2-en-1-imine 2d
Mp: 172–174°C; 1
H NMR (500 MHz, CDCl3): d 9.47 (s, 1H), 8.72
(dd, 1H, J ¼ 11.5, H-3), 8.42 (dd, 1H, J ¼ 8.5, H-6), 8.20 (s, 1H),
7.88 (m, 1H, J ¼ 8.0, H-60
), 7.82 (t, 1H, J ¼ 9.5, H-5), 7.79 (d, 1H,
J ¼ 12.5, Ha), 7.75 (d, 1H, J ¼ 9.5, H-50
), 7.44 (dd, 1H, J ¼ 11.0,
H-4), 6.92 (d, 1H, J ¼ 12.5, Hb), 3.88 (s, 3H); 13
C NMR (125 MHz,
DMSO-d6) d 162.5 (C––N), 153.5 (C1), 118.9 (C6), 134.4 (C5),
130.4 (C4), 150.2 (C3), 136.4 (C10
), 118.2 (C20
), 114.2 (C30
),
139.7 (C40
), 125.3 (C50
), 120.2 (C60
), 144.2 (vinylic), 133.2
(vinylic); IR (KBr) n (cmÀ1
): 3645, 3548, 2652, 1635, 1425, 1302,
1225, 805, 672; HRMS m/z: 300.0976 ([MþH])þ
; calcd.:
300.0984.
2-{(1E,2E)-3-[4-(Dimethylamino)phenyl]-N-hydroxyprop-2-
enimidoyl}phenol 2e
Mp: 158–160°C. 1
H NMR (500 MHz CDCl3): d 12.35 (s, 1H, OH),
9.52 (s, 1H), 7.04 (dd, 2H, J ¼ 10.5, H-30
and H-50
), 7.89 (d, 1H,
J ¼ 12.5, Ha), 7.79 (d, 1H, J ¼ 7.5, H-6), 7.72 (dd, 2H, J ¼ 10.0, H-
20
and H-60
), 7.46 (dd, 1H, J ¼ 8.0, H-4), 6.97 (t, 1H, J ¼ 9.5, H-3),
6.92 (d, 1H, J ¼ 12.5, Hb), 6.82 (dd, 1H, J ¼ 13.5, H-5); 13
C NMR
(100 MHz, DMSO-d6) d 160.3 (C––N), 127.2 (C1), 131.97 (C2 and
C6), 110.97 (C5 and C3), 154.33 (C4), 40.5 (N–CH3), 147.1
(vinylic), 127.2 (vinylic), 121.8 (C10
), 160.5 (C20
), 122.6 (C30
),
119.7 (C40
), 137.4 (C50
), 134.7 (C60
); IR (KBr) n (cmÀ1
): 3620,
3528, 2342, 1620, 1415, 1300, 1222, 817, 632; HRMS m/z:
283.1439 ([MþH])þ
; calcd.: 283.1447.
3-[(1E,2E)-N-Hydroxy-3-(2-methoxy-4-nitrophenyl)prop-2-
enimidoyl]naphthalen-2-ol 2f
Mp: 145–147°C; 1
H NMR (500 MHz, CDCl3): d 12.55 (s, 1H, OH),
9.70 (s, 1H), 8.17 (s, 1H), 8.09 (dd, 1H, J ¼ 10.5, H-40
), 7.85 (dd,
1H, J ¼ 9.0, H-80
), 7.74 (dd, 1H, J ¼ 8.0, H-70
), 7.58 (t, 1H,
J ¼ 10.0, H-50
), 7.57 (d, 1H, J ¼ 12.0, Hb), 7.52 (d, 1H, J ¼ 12.0,
Ha), 7.35 (dd, 1H, J ¼ 8.5, H-60
), 7.24 (dd, 1H, J ¼ 9.5, H-30
), 7.12
(d, 1H, J ¼ 7.5, H-30
), 3.72 (s, 3H); 13
C NMR (125 MHz, DMSO-d6)
d 162.5 (C––N), 126.1 (C10
), 160.8 (C20
), 120.1 (C30
), 130.4 (C40
),
126.4 (C50
), 130.5 (C60
), 127.2 (C70
), 130.4 (C80
), 129.9 (C90
),
140.2 (C100
), 132.2 (vinylic), 144.6 (vinylic), 58.6 (Me), 130.2
(C1), 120.8 (C2), 118.4 (C3), 129.4 (C4), 122.6 (C5), 120.2 (C6); IR
(KBr) n (cmÀ1
): 3650, 3255, 3024, 2842, 1692, 1600, 1557, 1443,
934, 720, 655, 538; HRMS m/z: 365.1131 ([MþH])þ
; calcd.:
365.1137.
3-[(1E,2E)-N-Hydroxy-3-(4-methoxy-3-nitrophenyl)prop-2-
enimidoyl]naphthalen-2-ol 2g
Mp: 132–134°C; 1
H NMR (500 MHz, CDCl3): d 12.05 (s, 1H, OH),
9.42 (s, 1H, OH), 8.20 (s, 1H), 7.89 (dd, 1H, J ¼ 10.5, H-5), 7.84
(dd, 1H, J ¼ 9.5, H-80
), 7.82 (dd, 1H, J ¼ 9.5, H-6), 7.74 (dd, 1H,
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
S. K. Radhakrishnan et al. Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 8
J ¼ 8.0, H-70
), 7.62 (d, 1H, J ¼ 13.0, Hb), 7.52 (t, 1H, J ¼ 10.0, H-
50
), 7.51 (dd, 1H, J ¼ 9.0, H-40
), 7.49 (d, 1H, J ¼ 13.0, Ha), 7.24
(dd, 1H, J ¼ 9.5, H-30
), 6.62 (dd, 1H, J ¼ 8.5, H-60
), 3.72 (s, 3H);
13
C NMR (125 MHz, DMSO-d6) d 159.8 (C––N), 119.2 (C10
), 120.4
(C20
), 148.4 (C30
), 150.2 (C40
), 123.4 (C50
), 128.8 (C60
), 122.4
(C70
), 129.4 (C80
), 130.4 (C90
), 140.25 (C100
), 126.9 (vinylic),
140.5 (vinylic), 122.8 (C1), 124.5 (C2), 140.2 (C3), 140.4 (C4),
116.82 (C5), 118.2 (C6), 58.7 (Me); IR (KBr) n (cmÀ1
): 3685, 3250,
2850, 2565, 2320, 1670, 1550, 1434, 1250, 935, 712, 535; HRMS
m/z: 365.1130 ([MþH])þ
; calcd.: 365.1137.
Pharmacology
Tyrosinase inhibition assay
The mushroom tyrosinase inhibition activity of chalcone
oxime compounds was measured using L-DOPA as sub-
strate [30]. Mushroom tyrosinase, L-DOPA, and tested samples
were prepared by dissolving in 50 mM Na2HPO4–NaH2PO4
buffer (pH 6.8). Reaction mixtures containing 50mL of 2 mmol/L
of L-DOPA, 50 mL of phosphate buffer, and 50 mL of different
concentrations (0.5, 1.0, 2.5, 5.0, and 10.0 mM) of tested
compounds were added in 96 well microtiter plates, followed
by the addition of 50 mL of 0.2 mg/mL of mushroom
tyrosinase. The assay mixture was incubated at 25°C for
30 min. Following incubation, the amount of DOPAchrome
produced in the reaction mixture was determined spectro-
photometrically at 492 nm using a Flex Station 3 microplate
reader. We determined the inhibition at 20 mM for all the
synthesized compounds. In addition, we determined the IC50,
that is, the concentration of compound that causes 50%
inhibition for kojic acid and the active inhibitors 2b and 2e. In
the present study, dose-dependent inhibition experiments
were performed in triplicate to determine the IC50 of test
compounds. The average results from three experiments are
shown.
Determination of the inhibition type of compounds 2b
and 2e on mushroom tyrosinase
Inhibitors were first dissolved in DMSO and used for the test
after a 30-fold dilution. The final concentration of DMSO in
the test solution was 3.33%. Mushroom tyrosinase (50 mL; 0.2
mg/mL) was incubated with 50 mL of various concentrations of
enzyme substrate (0.2–0.6 mM) and 50 mL of phosphate
buffer, and then 50 mL of different concentrations of tested
samples (0, 1.25, 5.0, and 20.0 mM) were simultaneously added
to the reaction mixtures. The measurement was performed in
triplicate for each concentration and averaged before further
calculation. The absorbance variations from these studies
were used to generate Lineweaver–Burk plots to determine
the inhibition type. The kinetic parameter (Km) of the
tyrosinase activity was calculated by linear regression from
Lineweaver–Burk plots. For the type of enzyme inhibition and
the inhibition constant (Ki) for an enzyme-inhibitor complex,
the mechanisms were analyzed by Dixon plot, which is a
graphical method [plot of 1/enzyme velocity (1/v) versus
inhibitor concentration (I) with varying concentrations of the
substrate].
In silico docking between tyrosinase and target
compounds
To further understand the binding modes of the most active
compounds with mushroom tyrosinase, molecular docking
studies of compounds 2b and 2e were performed using
Discovery Studio 4.5 (Accelrys, San Diego, CA, USA). To model
the tyrosinase structure, we used the crystal structure of
Agaricus bisporus tyrosinase (PDB ID: 2Y9X), A chain. From the
docking results, we checked for possible hydrogen-bonding
and non-bonding interactions with the amino acid residues.
Cell viability
Cell survival was quantified by a colorimetric assay that used 3-
(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
(MTT) and that measured mitochondrial activity in viable
cells. This method is based on the conversion of MTT (Sigma)
to MTT-formazan crystals by a mitochondrial enzyme. Briefly,
cells seeded at a density of 3 Â 104
/cell in a 48-well plate were
allowed to adhere overnight; the culture medium was then
replaced with fresh serum-free DMEM. MTT was freshly
prepared at 5 mg/mL in phosphate-buffered saline (PBS).
Aliquots of 500 mL of MTT stock solution were added to each
well, and the plate was incubated at 37°C for 4 h in a
humidified 5% CO2 incubator. After 4 h, the medium was
removed. To each well, 500 mL of EtOH-DMSO (solution of a
1:1 mixture) was added to dissolve the formazan. After
10 min, the optical density of each well was measured
spectrophotometrically using a 560 nm filter.
Assay of murine tyrosinase activity
Tyrosinase activity was estimated by measuring the rate of L-
DOPA oxidation. Cells were plated in 24-well dishes at a
density of 5 Â 104
cells/mL. B16 cells were incubated in the
presence or absence of 100 nM a-MSH, and then treated for
24 h with various concentrations of 2a or 2b (0–10 mM). Cells
were washed and lysed in 100 mL of 50 mM sodium phosphate
buffer (pH 6.5) containing 1% Triton X-100 (Sigma) and
0.1 mM PMSF (phenylmethylsulfonyl fluoride), and then
frozen at À80°C for 30 min. After thawing and mixing,
cellular extracts were clarified by centrifugation at 12000 rpm
for 30 min at 4°C. An 8 mL sample of the supernatant and 20 mL
of L-DOPA (2 mg/mL) were placed in a 96-well plate, and the
absorbance at 492 nm was read every 10 min for 1 h at 37°C
using an ELISA plate reader. The final activity is expressed as D
O.D. minÀ1
for each condition.
Determination of melanogenesis in B16 cells
Determination of melanin content of cells was done using a
modification of the method of Bilodeau et al. In the present
study, the amount of melanin was used as an index of
melanogenesis. B16 F10 cells (5 Â 104
) were transferred to 24-
well dishes and incubated in the presence or absence of
100 nM a-MSH. Cells were then incubated for 24 h with
various concentrations of 2a and 2b (0–10 mM). After washing
twice with PBS, samples were dissolved in 100 mL of 1 N NaOH.
The samples were then incubated at 60°C for 1 h and mixed to
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 9
solubilize the melanin. Absorbance at 405 nm was compared
with a standard curve of synthetic melanin.
The authors have declared no conflicts of interest.
References
[1] R. A. Spritz, V. J. Hearing, Adv. Hum. Genet. 1994, 22,
1–45.
[2] M. Brenner, V. J. Hearing, Photochem. Photobiol. 2008,
84, 539–549.
[3] K. Iozumi, G. E. Hoganson, R. Pennella, M. A. Everett,
B. B. Fuller, J. Invest. Dermatol. 1993, 100, 806–811.
[4] H. Ando, H. Kondoh, M. Ichihashi, V. J. Hearing, J. Invest.
Dermatol. 2007, 127, 751–61.
[5] H. M. Wang, C. Y. Chen, Z. H. Wen, Exp. Dermatol. 2011,
20, 242–248.
[6] A. B. Lerner, T. B. Fitzpatrick, E. Calkins,
W. H. Summerson, J Biol Chem. 1955, 187, 793–802.
[7] M. V. Martinez, J. R. Whitaker, Trends Food Sci. Tech.
1995, 6, 195–200.
[8] W. Yi, R. Cao, Eur. J. Med. Chem. 2010, 45, 639–646.
[9] Y.Xu,A.H.Stokes,W.M.Freeman,S.C. Kumer, B. A.Vogt,
K. E. Vrana, Mol. Brain Res. 1997, 45(1), 159–162.
[10] M. Ashida, P. Brey, Proc. Natl. Acad. Sci. USA 1995, 92,
10698–10702.
[11] S. Passi, P. M. Nazzaro, Br. J. Dermatol. 1981, 104,
659–665.
[12] M. Criton, V. Mellay-Hamon, Bioorg. Med. Chem. Lett.
2008, 18, 3607–3610.
[13] H. S. Baek, H. S. Rho, J. W. Yoo, S. M. Ahn, J. Y. Lee, J. Lee,
M. K. Kim, D. K. Kim, I. S. Chang, Bull. Kor. Chem. Soc.
2008, 29, 43–46.
[14] H. J. Lindner, S. Goettlicher, Acta Crystallogr. Sect. B
1969, 25, 832–842.
[15] S. K. Radhakrishnan, R. Shimmon Conn, A. T. Baker,
Bioorg. Med. Chem. Lett. 2015, 25, 1753–1756.
[16] C. Q. Meng, L. Ni, K. J. Worsencroft, Z. Ye,
M. D. Weingarten, J. E. Simpson, J. W. Skudlarek,
E. M. Marino, K. L. Suen, C. Kunsch, A. Souder,
R. B. Howard, C. L. Sundell, M. A. Wasserman,
J. A. Sikorski, J. Med. Chem. 2007, 50, 1304–1315.
[17] T. W. Yan, J. Q. Ya, L. Z. Ya, J. L. Yu, R. Bing, Q. Z. Yan,
R. Y. Meng, Q. J. Ai, L. Q. Jin, L. Z. Hai, RSC Adv. 2014, 61,
32263–32275.
[18] Y. Matoba, T. Kumagai, A. Yamamoto,
H. Yoshitsu, M. Sugiyama, J. Biol. Chem. 2008,
281, 8981–8990.
[19] H. Decker, F. Tuczek, Trends Biochem. Sci. 2000, 25,
392–397.
[20] M. Jimenez-Atienzar, J. Escribano, J. Cabanes, F. Gandıa-
Herrero, F. Garcıa-Carmona, Plant Physiol. Biochem.
2005, 43, 866–873.
[21] S. Ennes, R. Paschoalick, M. M. D. A. Alchorne,
J. Dermatol. Treat. 2000, 11, 173–179.
[22] P. G. Engasser, J. Am. Acad. Dermatol. 1984, 10,
1072–1073.
[23] A. Fisher, Cutis 1983, 31, 240–244, 250.
[24] C. Romaguera, F. Grimalt, Contact Dermatitis 1985, 12,
183.
[25] E. V. Curto, C. Kwong, H. Hermersdorfer, H. Glatt,
C. Santis, V. Virador, V. J. Hearing, T. P. Dooley, Biochem.
Pharmacol. 1999, 57, 663–672.
[26] H. Zhou, J. K. Kepa, D. Siegel, S. Miura, Y. Hiraki, D. Ross,
Mol. Pharmacol. 2009, 76, 579–587.
[27] V. Kahn, Pigment Cell Res. 1995, 8, 234–240.
[28] N. Fujimoto, H. Onodera, K. Mitsumori, T. Tamura,
S. Maruyama, A. Ito, Carcinogenesis 1999, 20,
1567–1571.
[29] Y. J. Zhu, L. Qiu, J. J. Zhou, H. Y. Guo, Y. H. Hu, Z. C. Li,
Q. Wang, Q. X. Chen, B. Liu, J. Enz. Inh. Med. Chem. 2010,
25, 798–803.
[30] J. K. No, D. Y. Soung, Y. J. Kim, K. H. Shim, Y. S. Jun,
S. H. Rhee, T. Yokozawa, H. Y. Chung, Life Sci. 1999, 65,
241–246.
Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10
S. K. Radhakrishnan et al. Archiv der Pharmazie
ARCHRCH PHARMHARM
ß 2015 WILEY-VCH Verlag GmbH  Co. KGaA, Weinheim www.archpharm.com 10

Weitere ähnliche Inhalte

Was ist angesagt?

Tetracylines and Macrolide antibiotics
Tetracylines and Macrolide antibioticsTetracylines and Macrolide antibiotics
Tetracylines and Macrolide antibioticsAkhil Nagar
 
Analog design medicinal chemistry
Analog design medicinal chemistryAnalog design medicinal chemistry
Analog design medicinal chemistryMohit umare
 
Drug discovery 30
Drug discovery 30Drug discovery 30
Drug discovery 30nitin lambe
 
Stability studies of proteins and peptides.
Stability studies of proteins and peptides.Stability studies of proteins and peptides.
Stability studies of proteins and peptides.SULABH910
 
Complexation and protein binding
Complexation and protein bindingComplexation and protein binding
Complexation and protein bindingPunitMadaan
 
Stability Of Peptides And Proteins
Stability Of Peptides And ProteinsStability Of Peptides And Proteins
Stability Of Peptides And ProteinsThilak Chandra
 
Fuctional group determination of drugs in biological activity.
Fuctional group determination of drugs in biological activity.Fuctional group determination of drugs in biological activity.
Fuctional group determination of drugs in biological activity.vishnu chinnamsetti
 
Molecular docking study_from_lunacridine_scopoleti(1)
Molecular docking study_from_lunacridine_scopoleti(1)Molecular docking study_from_lunacridine_scopoleti(1)
Molecular docking study_from_lunacridine_scopoleti(1)Adriani Hasyim
 
Catalysis Presentation
Catalysis PresentationCatalysis Presentation
Catalysis PresentationNisha Ahuja
 
Solution And Solid Phase Synthesis Publication
Solution  And Solid Phase Synthesis PublicationSolution  And Solid Phase Synthesis Publication
Solution And Solid Phase Synthesis Publicationadotse
 
Complexation and Protein Binding [Part-1] (Introduction and Classification an...
Complexation and Protein Binding [Part-1](Introduction and Classification an...Complexation and Protein Binding [Part-1](Introduction and Classification an...
Complexation and Protein Binding [Part-1] (Introduction and Classification an...Ms. Pooja Bhandare
 
C–H bond hydroxylation at non heme carboxylate-bridged diiron centers
C–H bond hydroxylation at non heme carboxylate-bridged diiron centersC–H bond hydroxylation at non heme carboxylate-bridged diiron centers
C–H bond hydroxylation at non heme carboxylate-bridged diiron centersDaniel Morton
 
APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...
APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...
APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...JamesSahn
 
Microbial Transformation of steroids
Microbial Transformation of steroidsMicrobial Transformation of steroids
Microbial Transformation of steroidsAnanyaVerma52
 
Medicinal Chemistry Basics
Medicinal Chemistry BasicsMedicinal Chemistry Basics
Medicinal Chemistry BasicsRahul Patil PhD
 

Was ist angesagt? (20)

Tetracylines and Macrolide antibiotics
Tetracylines and Macrolide antibioticsTetracylines and Macrolide antibiotics
Tetracylines and Macrolide antibiotics
 
Analog design medicinal chemistry
Analog design medicinal chemistryAnalog design medicinal chemistry
Analog design medicinal chemistry
 
Drug discovery 30
Drug discovery 30Drug discovery 30
Drug discovery 30
 
EPCI paper
EPCI paperEPCI paper
EPCI paper
 
Bioisosterism - Introduction
Bioisosterism - IntroductionBioisosterism - Introduction
Bioisosterism - Introduction
 
Stability studies of proteins and peptides.
Stability studies of proteins and peptides.Stability studies of proteins and peptides.
Stability studies of proteins and peptides.
 
Complexation and protein binding
Complexation and protein bindingComplexation and protein binding
Complexation and protein binding
 
Stability Of Peptides And Proteins
Stability Of Peptides And ProteinsStability Of Peptides And Proteins
Stability Of Peptides And Proteins
 
Macrolide antibiotics
Macrolide antibioticsMacrolide antibiotics
Macrolide antibiotics
 
Fuctional group determination of drugs in biological activity.
Fuctional group determination of drugs in biological activity.Fuctional group determination of drugs in biological activity.
Fuctional group determination of drugs in biological activity.
 
Molecular docking study_from_lunacridine_scopoleti(1)
Molecular docking study_from_lunacridine_scopoleti(1)Molecular docking study_from_lunacridine_scopoleti(1)
Molecular docking study_from_lunacridine_scopoleti(1)
 
Catalysis Presentation
Catalysis PresentationCatalysis Presentation
Catalysis Presentation
 
Solution And Solid Phase Synthesis Publication
Solution  And Solid Phase Synthesis PublicationSolution  And Solid Phase Synthesis Publication
Solution And Solid Phase Synthesis Publication
 
paper 2 published
paper 2 publishedpaper 2 published
paper 2 published
 
Complexation and Protein Binding [Part-1] (Introduction and Classification an...
Complexation and Protein Binding [Part-1](Introduction and Classification an...Complexation and Protein Binding [Part-1](Introduction and Classification an...
Complexation and Protein Binding [Part-1] (Introduction and Classification an...
 
C–H bond hydroxylation at non heme carboxylate-bridged diiron centers
C–H bond hydroxylation at non heme carboxylate-bridged diiron centersC–H bond hydroxylation at non heme carboxylate-bridged diiron centers
C–H bond hydroxylation at non heme carboxylate-bridged diiron centers
 
APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...
APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...
APPLICATIONS OF MULTICOMPONENT ASSEMBLY PROCESSES TO THE FACILE SYNTHESES OF ...
 
DPPH assay
DPPH assayDPPH assay
DPPH assay
 
Microbial Transformation of steroids
Microbial Transformation of steroidsMicrobial Transformation of steroids
Microbial Transformation of steroids
 
Medicinal Chemistry Basics
Medicinal Chemistry BasicsMedicinal Chemistry Basics
Medicinal Chemistry Basics
 

Andere mochten auch

環保單位持續加強市售玩具類商品所附乾電池查驗工作
環保單位持續加強市售玩具類商品所附乾電池查驗工作環保單位持續加強市售玩具類商品所附乾電池查驗工作
環保單位持續加強市售玩具類商品所附乾電池查驗工作epaslideshare
 
Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...
Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...
Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...Nii Korley Kortei
 
土壤及地下水年度盛會在南港展覽館登場
土壤及地下水年度盛會在南港展覽館登場土壤及地下水年度盛會在南港展覽館登場
土壤及地下水年度盛會在南港展覽館登場epaslideshare
 
INF20101 Equipo de Trabajo
INF20101 Equipo de TrabajoINF20101 Equipo de Trabajo
INF20101 Equipo de Trabajoguest92c025
 
2016 HCT Fan Optimisation
2016 HCT Fan Optimisation2016 HCT Fan Optimisation
2016 HCT Fan OptimisationMichael Dunn
 
UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR
UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR
UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR Annytha Panamá
 
Measured Perspective 3 Prompted
Measured Perspective 3 PromptedMeasured Perspective 3 Prompted
Measured Perspective 3 Prompteds2francetrip
 
Planometric Prompted
Planometric PromptedPlanometric Prompted
Planometric Prompteds2francetrip
 
1040721十大環境教育影片排行榜
1040721十大環境教育影片排行榜1040721十大環境教育影片排行榜
1040721十大環境教育影片排行榜epaslideshare
 

Andere mochten auch (18)

環保單位持續加強市售玩具類商品所附乾電池查驗工作
環保單位持續加強市售玩具類商品所附乾電池查驗工作環保單位持續加強市售玩具類商品所附乾電池查驗工作
環保單位持續加強市售玩具類商品所附乾電池查驗工作
 
Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...
Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...
Modeling the solar drying kinetics of gamma irradiation-pretreated oyster mus...
 
Inhumano
InhumanoInhumano
Inhumano
 
600.18 chameleon refund procedure
600.18 chameleon refund procedure600.18 chameleon refund procedure
600.18 chameleon refund procedure
 
土壤及地下水年度盛會在南港展覽館登場
土壤及地下水年度盛會在南港展覽館登場土壤及地下水年度盛會在南港展覽館登場
土壤及地下水年度盛會在南港展覽館登場
 
Respiraprofundo
RespiraprofundoRespiraprofundo
Respiraprofundo
 
INF20101 Equipo de Trabajo
INF20101 Equipo de TrabajoINF20101 Equipo de Trabajo
INF20101 Equipo de Trabajo
 
香港六合彩
香港六合彩香港六合彩
香港六合彩
 
Tesis
TesisTesis
Tesis
 
2016 HCT Fan Optimisation
2016 HCT Fan Optimisation2016 HCT Fan Optimisation
2016 HCT Fan Optimisation
 
UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR
UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR
UNIDAD EDUCATIVA REPÚBLICA DEL ECUADOR
 
Measured Perspective 3 Prompted
Measured Perspective 3 PromptedMeasured Perspective 3 Prompted
Measured Perspective 3 Prompted
 
Els Estats Units, un immens potencial per l'empresa catalana / ACC1Ó
Els Estats Units, un immens potencial per l'empresa catalana / ACC1ÓEls Estats Units, un immens potencial per l'empresa catalana / ACC1Ó
Els Estats Units, un immens potencial per l'empresa catalana / ACC1Ó
 
Planometric Prompted
Planometric PromptedPlanometric Prompted
Planometric Prompted
 
Anglicismos e Pidgins
Anglicismos e PidginsAnglicismos e Pidgins
Anglicismos e Pidgins
 
Barrera
BarreraBarrera
Barrera
 
1040721十大環境教育影片排行榜
1040721十大環境教育影片排行榜1040721十大環境教育影片排行榜
1040721十大環境教育影片排行榜
 
Projeto Grafico | Identidade CEAGESP
Projeto Grafico | Identidade CEAGESPProjeto Grafico | Identidade CEAGESP
Projeto Grafico | Identidade CEAGESP
 

Ähnlich wie paper 6 published

Atividade antioxidante2
Atividade antioxidante2Atividade antioxidante2
Atividade antioxidante2Sarah Silveira
 
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...Fernando Hernandez Borja
 
Arsenic and Bladder Cancer
Arsenic and Bladder Cancer Arsenic and Bladder Cancer
Arsenic and Bladder Cancer Mezbahul Haque
 
1-s2.0-S1011134414003510-main
1-s2.0-S1011134414003510-main1-s2.0-S1011134414003510-main
1-s2.0-S1011134414003510-mainAvinash Gandi
 
17064-9fQK1439202911-
17064-9fQK1439202911-17064-9fQK1439202911-
17064-9fQK1439202911-rajkutty1983
 
Antiaging Effect of Leaves of Different Extract Salvia Splendens
Antiaging Effect of Leaves of Different Extract Salvia SplendensAntiaging Effect of Leaves of Different Extract Salvia Splendens
Antiaging Effect of Leaves of Different Extract Salvia SplendensBRNSS Publication Hub
 
Evaluation of salicin isolated from salix subserrata as a radioprotector
Evaluation of salicin isolated from salix subserrata as a radioprotectorEvaluation of salicin isolated from salix subserrata as a radioprotector
Evaluation of salicin isolated from salix subserrata as a radioprotectorRam Sahu
 
Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...
Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...
Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...Editor IJCATR
 
Thesis extended abstract Raissa
Thesis extended abstract RaissaThesis extended abstract Raissa
Thesis extended abstract RaissaRaissa Melina
 
Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorrkkoiri
 
Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorrkkoiri
 

Ähnlich wie paper 6 published (20)

paper 7 published
paper 7 publishedpaper 7 published
paper 7 published
 
paper 5
paper 5paper 5
paper 5
 
paper 4
paper 4paper 4
paper 4
 
Chalcones
ChalconesChalcones
Chalcones
 
Atividade antioxidante2
Atividade antioxidante2Atividade antioxidante2
Atividade antioxidante2
 
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
69. synthesis, ex vivo and in silico studies of 3 cyano-2-pyridone derivative...
 
Gallery (2).pptx
Gallery (2).pptxGallery (2).pptx
Gallery (2).pptx
 
5. C.J. Patil Article.pdf
5. C.J. Patil Article.pdf5. C.J. Patil Article.pdf
5. C.J. Patil Article.pdf
 
Arsenic and Bladder Cancer
Arsenic and Bladder Cancer Arsenic and Bladder Cancer
Arsenic and Bladder Cancer
 
1-s2.0-S1011134414003510-main
1-s2.0-S1011134414003510-main1-s2.0-S1011134414003510-main
1-s2.0-S1011134414003510-main
 
BMC ASHWINI
BMC ASHWINIBMC ASHWINI
BMC ASHWINI
 
INVENTI-LAVANYA
INVENTI-LAVANYAINVENTI-LAVANYA
INVENTI-LAVANYA
 
17064-9fQK1439202911-
17064-9fQK1439202911-17064-9fQK1439202911-
17064-9fQK1439202911-
 
Antiaging Effect of Leaves of Different Extract Salvia Splendens
Antiaging Effect of Leaves of Different Extract Salvia SplendensAntiaging Effect of Leaves of Different Extract Salvia Splendens
Antiaging Effect of Leaves of Different Extract Salvia Splendens
 
J med chem-2
J med chem-2J med chem-2
J med chem-2
 
Evaluation of salicin isolated from salix subserrata as a radioprotector
Evaluation of salicin isolated from salix subserrata as a radioprotectorEvaluation of salicin isolated from salix subserrata as a radioprotector
Evaluation of salicin isolated from salix subserrata as a radioprotector
 
Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...
Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...
Phytochemical Studies on Linum Usitatissimum Seeds and the Nanoformulation of...
 
Thesis extended abstract Raissa
Thesis extended abstract RaissaThesis extended abstract Raissa
Thesis extended abstract Raissa
 
Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulator
 
Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulator
 

paper 6 published

  • 1. Full Paper Evaluation of Novel Chalcone Oximes as Inhibitors of Tyrosinase and Melanin Formation in B16 Cells Sini K. Radhakrishnan, Ronald G. Shimmon, Costa Conn, and Anthony T. Baker School of Chemistry and Forensic Science, University of Technology Sydney, Ultimo, NSW, Australia A series of hydroxy-substituted chalcone oxime derivatives were synthesized. These compounds were then evaluated for their inhibitory activities on tyrosinase and melanogenesis in murine B16F10 melanoma cells. The structures of the synthesized compounds were confirmed by 1 H NMR, 13 C NMR, FTIR, and HRMS. Two of the compounds exhibited much higher tyrosinase inhibitory activities (IC50 values of 4.77 and 7.89 mM, respectively) than the positive control, kojic acid (IC50: 22.25 mM). Kinetic studies revealed them to act as competitive tyrosinase inhibitors with their Ki values of 5.25 and 8.33 mM, respectively. Both the compounds inhibited melanin production and tyrosinase activity in B16 cells. Docking results confirmed that the active inhibitors strongly interacted with the mushroom tyrosinase residues. Keywords: Chalcone oxime / Competitive / Docking / Tyrosinase inhibition Received: August 24, 2015; Revised: October 14, 2015; Accepted: October 19, 2015 DOI 10.1002/ardp.201500298 :Additional supporting information may be found in the online version of this article at the publisher’s web-site. Introduction Melanogenesis is the process by which melanin, a polymeric dark pigment, is produced and subsequently distributed by melanocytes within the skin and hair follicles [1]. Melanin plays a crucial role against skin photocarcinogenesis [2]. Excess production of melanin from melanocytes can lead to hyperpigmentation, which can manifest as melasma, cafe au lait macules, ephelides (freckles), and dark spots [3–5]. The development and screening of potent inhibitors of tyrosinase is therefore of particular interest to the cosmetic industry. Tyrosinase is the key enzyme involved in melanin biosyn- thesis responsible for the hydroxylation of L-tyrosine to L-DOPA (L‐3,4‐dihydroxyphenylalanine) and oxidation of L-DOPA to DOPA quinone [6]. Previous reports confirmed that tyrosinase was not only involved in melanizing in animals, but also was one of the main causes for loss of quality in fruits and vegetables during post-harvest handling and processing, leading to faster degradation and shorter shelf life [7, 8]. Additionally, tyrosinase may play a significant role in neuromelanin formation in the human brain and could be responsible for the neurodegeneration associated with Parkinson’s disease [9]. Also, in insects, tyrosinase is uniquely associated with three different biochemical processes, includ- ing sclerotization of cuticle, defensive encapsulation and melanization of foreign organisms, and wound healing [10]. Most melanin-biosynthesis inhibitors are phenol or catechol analogs, which are structurally similar to the tyrosinase substrates, tyrosine, or L-DOPA [11]. Besides, our interest for chelator agents led us to study the hydroxamic acid group. Hydroxamate molecules, one of the major classes of naturally occurring metal complexing agents, have been thoroughly studied as ligands for different metal ions such as Fe(III), Zn(II), and Cu(II) [12, 13]. The chelation involves the oxygen belonging to the ––N–OH group [14]. Previously, we have reported the tyrosinase inhibitory potential of novel aza- chalcone compounds [15]. The presence in chalcones of a conjugated double bond and a completely delocalized p electron system reduces their redox potentials and makes them susceptible to electron transfer reactions. The a–b unsaturated bond enables the chalcone to act as a Michael acceptor for nucleophilic species including glutathione (GSH) Correspondence: Dr. Sini K. Radhakrishnan, School of Chemistry and Forensic Science, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia. E-mail: Sini.KaranayilRadhakrishnan@student.uts.edu.au Fax: þ61295142000 Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 1
  • 2. or cysteine residues on proteins [16]. Previous studies have indicated the use of chalcone oximes as anticancer agents [17]. This prompted us to further investigate the effect of chalcone oximes on melanin formation in murine B16F10 cells. From a structural perspective, tyrosinase has two copper ions in its active site which play a vital role in its catalytic activity. Each of the copper ions is coordinated by three histidines in the protein matrix [18]. At the active site of tyrosinase, a dioxygen molecule binds in a side-on coordina- tion mode between two copper ions. The copper atoms participate directly in hydroxylation of monophenols to diphenols (cresolase activity) and in the oxidation of o-diphenols to o-quinones (catechol oxidase activity) that enhance the production of the brown color [19, 20]. Inhibition of the enzyme tyrosinase has been a major motivation for researchers, and several inhibitors are currently used as cosmetic additives and medicinal products for the treatment of hyperpigmentation. Although hydroquinone has been the mainstay treatment for hyperpigmentation, its clinical poten- tial has been complicated by a number of adverse reactions including contact dermatitis, irritation, transient erythema, burning, prickling sensation, leukoderma, chestnut spots on the nails, hypochromia, and ochronosis [21–25]. Moreover, hydroquinone is potentially mutagenic to mammalian cells. Hydroquinone and its derivative arbutin are both catabolized to benzene metabolites with the potential for bone marrow toxicity [26]. Another alternative, kojic acid is currently applied as a cosmetic skin whitener and food additive to prevent enzymatic browning [27]. However, its use in cosmetics has been limited, because of its instability during storage [28]. Further investigation of potential tyrosinase inhibitors that could be applied during food processing to curb enzymatic browning and for the treatment of hyperpig- mentation disorders is warranted. Results and discussion Chemistry and inhibition kinetics In the current study, we report the successful identification of several hydroxy-substituted chalcone oxime compounds with improved tyrosinase inhibition. A series of hydroxy-substi- tuted chalcone compounds were synthesized by base cata- lyzed Claisen–Schmidt condensation of an aldehyde and an appropriate ketone in a polar solvent like methanol. These compounds were then subsequently converted to their oxime derivatives by reaction with hydroxylamine hydrochlo- ride (Scheme 1). The structures of the compounds synthesized were confirmed by 1 H NMR, 13 C NMR, FTIR, and HRMS. Assays were performed with L-DOPA as the substrate, using kojic acid, a well-known effective tyrosinase inhibitor as the positive control. The inhibitory activities of the synthesized oxime compounds 2a–g were compared with the reference compound kojic acid (Fig. 1). In terms of the structure–activity relationships, compound 2b (2-[(1E,2E)-N-hydroxy-3-(2-methoxy-4-nitrophenyl)prop-2- enimidoyl]phenol) exhibited the most potent tyrosinase inhibitory activity with inhibition of 80.19%. This could be accounted to the presence of a strongly activating ortho- methoxy substituent and a meta directing nitro substituent. The presence of a strong electron releasing group in the ortho position (–OMe) and an electron withdrawing group (–NO2) in the para position modulates the electronic structure of ring B significantly. It is equally likely for the oxime moiety to coordinate with the copper atoms present in the tyrosinase active site. Also, shifting the methoxy substituent to the para position in ring B led to a decrease in tyrosinase inhibitory activity as exhibited by compound 2d. Replacement of the phenyl ring with a pyridine ring brought about a decline in tyrosinase inhibition (60.6%) as seen with compound 2a. This shows that ring A is not oriented such that the pyridine nitrogen can bind the copper center when compared to kojic acid. Compound 2e with an electron donating para-dimethyl amino group on ring B showed an inhibition of 72.12% at 20 mM. The inhibition of both compounds 2b and 2e was seen to be greater than the positive control kojic acid which exhibited 47.67% inhibition at 20 mM. Compounds 2a, 2c, and 2d showed significant tyrosinase inhibitions. Replacement of phenyl ring with a naphthyl ring gave compound 2f with moderate tyrosinase inhibition whereas compound 2g showed low inhibitory activity (50.6%) against mushroom tyrosinase. In particular, compounds 2b and 2e exhibited the greatest inhibition of L-DOPA oxidase activity of mushroom tyrosinase. In this study, we investigated in depth the tyrosinase inhibitory activities of compounds 2b and 2e. As shown in Table 1, compounds 2b, 2e, and kojic acid inhibited mushroom tyrosinase activity in a concentration-dependent manner. Both the compounds 2b and 2e had better inhibitory activities with IC50 values of 4.77 and 7.89mM, respectively, than the reference compound kojic acid (IC50: 22.25 mM). To explore the mechanism, we examined the kinetics of tyrosinase inhibition in the presence of the most active compounds 2b and 2e. The data are shown in Table 2. As the concentrations of active inhibitors 2b and 2e increased, Km values gradually increased, but Vmax values did not change, thereby indicating that the inhibitors act as competitive inhibitors of mushroom tyrosinase (Fig. 2). Dixon plots gave a family of straight lines passing through the same point at the second quadrant, giving the inhibition constant (Ki) (Fig. 3). The Ki value estimated from this Dixon plot was 5.25 and 8.33mM for the compounds 2b and 2e, respectively. Kinetic results indicate that compounds 2b and 2e might inhibit the tyrosinase activity by binding to the copper containing active site region. To confirm this, we have further simulated docking of these active compounds with mushroom tyrosinase. Docking results The Accelrys Discovery Studio 4.5 suite was utilized to simulate binding between mushroom tyrosinase and active tyrosinase inhibitors 2band2e. Tomodelthetyrosinasestructure,weused the crystal structure of Agaricus bisporus (mushroom) Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 S. K. Radhakrishnan et al. Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 2
  • 3. tyrosinase (PDB ID: 2Y9X) A chain. Among all the compounds synthesized, compounds 2b and 2e had the highest docking scoreswhencomparedwiththestandard,kojicacid(dockscore: À10.29 kcal/mol). Docking results showed that compound 2b hadbetterbindingcapability(dockscore:À28.94 kcal/mol)with mushroomtyrosinaseascomparedtocompound2e(dockscore: À27.45 kcal/mol) (Fig. 4). The greater inhibitory efficacy of compound 2b is attributed to chelation of the oxime oxygen atomwiththecopperion(Cu401)intheactivesiteofmushroom tyrosinase (2.29 Å). We searched for tyrosinase residues that might bind to compounds 2b and 2e, and found that the most important binding residues for interaction are His65, His85, Val248, Ala286, His244, Asn260, Val283, Met280, Ser282, His259, Phe292, and Glu322 for both compounds 2b and 2e. Strong hydrogen bonding with His61, His85, and Val283 was seen for compound 2b whereas compound 2e exhibited hydrogen bonding interactions with Asn260, Glu322, and His259. Both the active compounds showed hydrophobic p–p stacking interactions with His263 and T-shaped edge to face aryl–aryl interactions with Phe264 and His244. In vitro cytotoxic studies Our work was further extended to investigate the effect of novel chalcone oxime compounds 2b and 2e on the tyrosinase activity and melanin levels in B16F10 melanoma cells. The cytotoxic effects of 2b and 2e were estimated by measuring cell viability, where no significant cytotoxic effect Scheme 1. General method for the synthesis of hydroxy-substituted chalcones and their oxime derivatives. Reagents and conditions: (a) MeOH, NaOH, 0°C, 24 h; (b) NH2OH.HCl. 0 10 20 30 40 50 60 70 80 90 2g2f2e2d2c2b2aKA TyrosinaseinhibiƟon(%,20μM) Compound *** *** *** *** *** *** * Figure 1. Inhibitory effects of chalcone oximes on mushroom tyrosinase activity. Values represent means Æ SE of three determinations. Inhibitory concentration is 20 mM. Ã p 0.05 and ÃÃÃ p 0.001 compared to kojic acid. KA, kojic acid. Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 3
  • 4. was found at any of the concentrations tested. At doses of 1.0, 10.0, 50.0, and 100.0 mM of compounds 2b or 2e for 48 h, cell viability was 118, 92.2, 78.4, and 72.6% for compound 2b and 89, 82, 76.4, and 64% for compound 2e, respectively, compared with the control (Fig. 5). Thus, neither compound 2b nor 2e were cytotoxic to B16 cells in the concentration range of 1.0–100.0 mM. We then ascertained the inhibitory effects of compounds 2b and 2e on melanogenesis by quantifying the melanin content of B16 cells treated with compounds 2b and 2e (Fig. 6). The melanin content of B16 cells after treatment with compound 2b in the presence of 100 nM a-melanocyte-stimulating hormone (a-MSH) de- creased dose-dependently, showing 215.25% at 1.0 mM, 175.16% at 5.0 mM, and 112.0% at 10.0 mM, compared with the 100 nM a-MSH-only treated group (225.22%) and the control group (100%). Similarly, cells treated with compound 2e exhibited melanin contents of 235.17% at 1.0 mM, 215.55% at 5.0 mM, and 165.35% at 10.0 mM compared with 100 nM a-MSH-only-treated group (225.22%) and the control group (100%). Finally, we determined the inhibitory effects of compounds 2b and 2e on tyrosinase activity of B16 cells treated with 100 nm a-MSH. After 24 h incubation with 2b and 2e, tyrosinase activities were 145.91% at 1.0 mM, 114.52% at 5.0 mM, and 108.77% at 10.0 mM of 2b (Fig. 7), compared with the 100 nM a-MSH-only treated group (225.25%) and the control group (100%). Similarly, cells treated with compound 2e exhibited melanin contents of 160.45% at 1.0 mM, 126.5% at 5.0 mM, and 111.22% at 10.0 mM compared with 100 nM a-MSH-only-treated group (225.25%) and the control group (100%). The inhibition of compounds 2b and 2e on murine tyrosinase activity was greater than the reference compound kojic acid. The results establish that compounds 2b and 2e suppress melanin biosynthesis through the inhibition of tyrosinase. Conclusion To summarize, among hydroxy-substituted chalcone oxime compounds, compounds 2b and 2e were found to be more potent than the positive reference compound kojic acid. Kinetic studies identified both the compounds to be competitive inhibitors of mushroom tyrosinase. Docking simulation studies determined the possible residues of tyrosinase that could play a key role in binding the inhibitor molecules. In the B16 cell culture system, tyrosinase activity and melanin levels were significantly reduced in the presence of compounds 2b and 2e, suggesting therefore inhibition of melanin biosynthesis to be attributed to tyrosinase inhibition. The structures of the non-cyclic moieties of our molecules have similar features to those of hydroxamic acids and hydroxamates which are good chelating agents. Tyrosinase inhibition of compounds 2b and 2e depended on the competency of the inhibitor compounds to block access by any substrate molecule in the active site of the enzyme. Strong coordination with the copper metal, significant hydrophobic and p–p stacking interactions contributed to effective Table 2. Kinetic analysis of compounds 2b and 2e. Compound Type of inhibition Vmax (mM/min) Ki (mM)a) 2b Competitive 0.045 5.25 2e Competitive 0.023 8.33 a) Data are represented as mean values of three independent tests. Ki is the (inhibitor constant). Vmax is the maximum reaction velocity. Table 1. Inhibitory effects of kojic acid, 2b, and 2e on mushroom tyrosinase activity. Sample Concentration (mM) Inhibition (%) Average of inhibition (%) IC50 (mM)a) Kojic acid 1.25 2.19 3.90 2.65 2.91 22.25 Æ 0.22 5.00 29.22 28.53 30.22 29.32 20.00 49.26 47.54 46.23 47.67 2b 1.25 43.20 44.17 42.65 43.34 4.77 Æ 0.35 5.00 49.60 50.25 57.44 52.43 20.00 80.60 78.77 81.22 80.19 2e 1.25 39.36 34.25 35.50 36.37 7.89 Æ 0.15 5.00 45.22 47.21 44.65 45.69 20.00 72.50 70.23 73.65 72.12 a) Fifty percent inhibitory concentration (IC50). Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 S. K. Radhakrishnan et al. Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 4
  • 5. tyrosinase inhibition. Also, oximes, popular in supramolecular chemistry as strong hydrogen bond donors, were involved in hydrogen bond interactions with the amino acid residues of the enzyme [29]. This, in turn, helped the compound to have a better fit to the catalytic pocket of the enzyme tyrosinase. The current study portrays chalcone oximes as promising candi- dates to be explored as depigmentation agents. Experimental Chemistry Chemical reagents and instruments Melting points (mp) were determined with WRS-1B melting point apparatus and the thermometer was uncorrected. NMR spectra were recorded on Agilent 500 spectrometer at 25°C in CDCl3 or DMSO-d6. All chemical shifts (d) are quoted in parts per million downfield from TMS and coupling constants (J) are given in Hertz. Abbreviations used in the splitting pattern were as follows: s ¼ singlet, d ¼ doublet, t ¼ triplet, m ¼ mul- tiplet. HRMS spectra were recorded using the Agilent Technologies 6520 LC/MS-QTOF. All reactions were monitored by TLC (Merck Kieselgel 60 F254) and the spots were visualized under UV light. Infrared (IR) spectra were recorded on Thermo Scientific NICOLET 6700 FT-IR spectrometer. Tyrosinase, L-3,4- dihydroxyphenylalanine (L-DOPA) and kojic acid were pur- chased from Sigma–Aldrich Chemical Co. General method for the synthesis of hydroxy chalcone derivatives (1a–g) To a stirred solution of the appropriate ketone (1 mM) and a substituted aldehyde (1 mM) in 25 mL methanol, was added pulverized NaOH (2 mM) and the mixture was stirred at room temperature for 24–36 h. The reaction was monitored by TLC using n-hexane/ethyl acetate (7:3) as mobile phase. The reaction mixture was cooled to 0°C (ice-water bath) and acidified with HCl (10% v/v aqueous solution) to afford total precipitation of the compounds. In most cases, a yellow precipitate was formed, which was filtered and washed with 10% aqueous HCl solution. In the cases where an orange oil was formed, the mixture was extracted with CH2Cl2, the extracts were dried (Na2SO4), and the solvent was evaporated to give the respective chalcone (1a–g). (2E)-1-(2-Hydroxyphenyl)-3-(2-methoxy-4-nitrophenyl)- prop-2-en-1-one 1a Mp: 123–125°C; 1 H NMR (500 MHz, CDCl3): d 12.22 (s, 1H, OH), 8.22 (s, 1H), 7.95 (d, 1H, J ¼ 14.0, Hb,), 7.86 (m, 1H, J ¼ 9.5, H-6), 7.80 (d, 1H, J ¼ 14.0, Ha), 7.75 (d, 1H, J ¼ 10.5, H-60 ), 7.69 (d, 1H, J ¼ 8.5, H-5), 7.59 (dd, 1H, J ¼ 8.5, H-40 ), 7.02 (dd, 1H, J ¼ 9.0, H- -200 -100 0 100 200 300 400 -4 -3 -2 -1 0 1 2 3 4 5 6 1/V(µM/min)-1 1/S (mM/L) 20 µM 5 µM 1.25 µM No inhibitor 2b -200 -100 0 100 200 300 400 -4 -2 0 2 4 6 1/V(mM/min)-1 1/S (mM/L) 20 µM 5 µM 1.25 µM No inhibitor 2e Figure 2. Lineweaver–Burk plot for inhibi- tion of compounds 2b and 2e on mushroom tyrosinase. Data were obtained as mean values of 1/V, the inverse of the absorbance increase at a wavelength of 492 nm/min of three independent tests with different con- centrations of L-DOPA as a substrate. The concentrations of compounds 2b and 2e from top to bottom are 20, 5, 1.25, and 0 mM, respectively. Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 5
  • 6. 30 ), 6.97 (m, 1H, J ¼ 10.0, H-50 ), 3.82 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 194.2 (C––O), 130.2 (C60 ), 119.33 (C10 ), 118.2 (C50 ), 137.0 (C40 ), 118.4 (C30 ), 162.9 (C20 ), 128.2 (C1), 115.2 (C2), 112.2 (C3), 137.5 (C4), 122.4 (C5), 116.5 (C6), 125.5 (vinylic), 140.8 (vinylic); IR (KBr) n (cmÀ1 ): 3250, 3345, 3015, 2910, 2865, 2700, 1702, 1682, 1552, 1465, 1305, 979, 735, 680, 575; MS (ESI): 270.1 ([MþH])þ . (2E)-3-(2-Methoxy-4-nitrophenyl)-1-(pyridin-2-yl)prop-2- en-1-one 1b Mp: 126–128°C; 1 H NMR (500 MHz, CDCl3): d 8.70 (dd, 1H, J ¼ 10.0, H-3), 8.32 (dd, 1H, J ¼ 8.5, H-6), 7.90 (d, 1H, J ¼ 12.0, Ha), 8.17 (s, 1H), 7.94 (m, 1H, J ¼ 8.0, H-60 ), 7.87 (t, 1H, J ¼ 9.0, H-5), 7.79 (d, 1H, J ¼ 10.5, H-50 ), 7.40 (m, 1H, J ¼ 10.5, H-4), 6.95 (d, 1H, J ¼ 12.0, Hb), 3.86 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 195.5 (C––O), 155.2 (C1), 120.9 (C6), 136.2 (C5), 129.4 (C4), 150.2 (C3), 134.0 (C10 ), 116.2 (C20 ), 112.2 (C30 ), 138.2 (C40 ), 125.3 (C50 ), 118.2 (C60 ), 140.2 (vinylic), 129.2 (vinylic); IR (KBr) n (cmÀ1 ): 3015, 2952, 1669, 1640, 1405, 1355, 1265, 1175, 805, 672; MS (ESI): 285.1 ([MþH])þ . (2E)-1-(2-Hydroxyphenyl)-3-(4-methoxy-3-nitrophenyl)- prop-2-en-1-one 1c Mp: 110–112°C; 1 H NMR (500 MHz, CDCl3): d 12.47 (s, 1H, OH), 8.10 (s, 1H), 7.86 (d, 1H, J ¼ 12.5, Hb), 7.74 (d, 1H, J ¼ 10.0, H-60 ), 7.72 (d, 1H, J ¼ 12.5, Ha), 7.65 (d, 1H, J ¼ 9.5, H-5), 7.54 (d, 1H, J ¼ 8.0, H-6), 7.49 (t, 1H, J ¼ 8.5, H-40 ), 6.92 (dd, 1H, J ¼ 9.5, H- 30 ), 6.85 (dd, 1H, J ¼ 9.0, H-50 ), 3.86 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 192.2 (C––O), 130.5 (C60 ), 119.2 (C10 ), 118.2 (C50 ), 136.7 (C40 ), 118.2 (C30 ), 161.9 (C20 ), 126.9 (C1), 120.5 (C2), 137.2 (C3), 147.5 (C4), 112.4 (C5), 114.2 (C6), 56.6 (Me), 122.4 (vinylic), 141.8 (vinylic); IR (KBr) n (cmÀ1 ): 3200, 3070, 2914, 2864, 2720, 1686, 1578, 1550, 1468, 1349, 970, 720, 580; MS (ESI): 270.1 ([MþH])þ . (2E)-3-(4-Methoxy-3-nitrophenyl)-1-(pyridin-2-yl)prop-2- en-1-one 1d Mp: 116–118°C; 1 H NMR (500 MHz, CDCl3): d 8.75 (dd, 1H, J ¼ 9.5, H-30 ), 8.25 (dd, 1H, J ¼ 8.5, H-60 ), 8.17 (s, 1H), 8.12 (s, -40 -30 -20 -10 0 10 20 30 40 -40 -30 -20 -10 0 10 20 30 1/v(µM/min)-1 [inhibitor] µM 2e -40 -30 -20 -10 0 10 20 30 40 50 -40 -30 -20 -10 0 10 20 30 1/v(µM/min)-1 [inhibitor] µM 2b Figure 3. Dixon plot for the inhibitory effect of compounds 2b and 2e on L-DOPA oxidation catalyzed by mushroom tyrosinase. The inhibitor concentrations were 0, 10, and 20 mM, res- pectively. The L-DOPA concentrations were 200, 400, and 600 mM. Figure 4. Docking results of compounds 2b and 2e in the tyrosinase catalytic pocket. Ligands 2b and 2e are displayed as ball and stick whereas the core amino acid residues are displayed as stick. The green-dotted lines show the hydrogen bond interactions and the purple lines show the non-bonding interactions. The ochre balls represent the copper ions. Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 S. K. Radhakrishnan et al. Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 6
  • 7. 1H), 7.89 (d, 1H, J ¼ 13.5, Ha), 7.86 (t, 1H, J ¼ 9.5, H-50 ), 7.69 (d, 1H, J ¼ 9.0, H-5), 7.57 (d, 1H, J ¼ 8.5, H-6), 7.47 (dd, 1H, J ¼ 11.0, H-40 ), 6.94 (d, 1H, J ¼ 13.5, Hb), 3.90 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 162.2 (C––N), 154.4 (C1), 121.2 (C6), 134.2 (C5), 130.4 (C4), 148.2 (C3), 135.4 (C10 ), 118.2 (C20 ), 114.2 (C30 ), 140.7 (C40 ), 125.3 (C50 ), 120.4 (C60 ), 139.2 (vinylic), 129.7 (vinylic); IR (KBr) n (cmÀ1 ): 3020, 2955, 1670, 1640, 1435, 1350, 1252, 1175, 805, 672; MS (ESI): 285.1 ([MþH])þ . (2E)-3-[4-(Dimethyl amino)phenyl]-1-(2-hydroxyphenyl)- prop-2-en-1-one 1e M.p: 108–110°C; 1 H NMR (500 MHz CDCl3): d 12.26 (s, 1H, OH), 8.06 (d, 1H, J ¼ 11.5, Hb), 7.85 (d, 1H, J ¼ 11.5, Ha), 7.72 (d, 1H, J ¼ 5.5, H-6), 7.68 (dd, 2H, J ¼ 8.0, H-20 and H-60 ), 7.49 (dd, 1H, J ¼ 8.5, H-4), 6.92 (t, 1H, J ¼ 9.0, H-3), 6.89 (dd, 1H, J ¼ 15.0, H- 5), 6.64 (dd, 2H, J ¼ 9.0, H-30 and H-50 ); 13 C NMR (100 MHz, DMSO-d6) d 190.3 (C––O), 125.1 (C1), 131.9 (C2 and C6), 110.9 (C5 and C3), 154.3 (C4), 40.0 (N–CH3), 145.3 (vinylic), 125.1 (vinylic), 119.7 (C10 ), 162.3 (C20 ), 118.1 (C30 ), 118.3 (C40 ), 136.4 (C50 ), 130.9 (C60 ); IR (KBr) n (cmÀ1 ): 3420, 1710, 1640, 1335, 813, 680, 757, 680; MS (ESI): 268.2 ([MþH])þ . (2E)-1-(3-Hydroxynaphthalen-2-yl)-3-(2-methoxy-4- nitrophenyl)prop-2-en-1-one 1f Mp: 115–117°C; 1 H NMR (500 MHz, CDCl3): d 12.69 (s, 1H, OH), 8.37 (dd, 1H, J ¼ 10.5, H-50 ), 8.20 (dd, 1H, J ¼ 9.0, H-100 ), 7.68 (dd, 1H, J ¼ 9.5, H-80 ), 7.60 (dd, 1H, J ¼ 8.5, H-60 ), 7.57 (d, 1H, J ¼ 12.0, Hb), 7.52 (d, 1H, J ¼ 12.0, Ha), 7.40 (m, 1H, J ¼ 10.0, H- 70 ), 7.25 (m, 1H, J ¼ 8.5, H-30 ); 13 C NMR (125 MHz, DMSO-d6) d 194.5 (C––O), 125.4 (C10 ), 159.8 (C20 ), 119.2 (C30 ), 130.2 (C40 ), 125.4 (C50 ), 132.5 (C60 ), 126.4 (C70 ), 132.4 (C80 ), 130.2 (C90 ), 144.2 (C100 ), 130.2 (vinylic), 149.8 (vinylic), 55.6 (Me), 130.2 (C1), 119.8 (C2), 117.4 (C3), 130.4 (C4), 124.7 (C5), 119.5 (C6); IR (KBr) n (cmÀ1 ): 3350, 3249, 3017, 2850, 2650, 1685, 1605, 1552, 1450, 1325, 975, 728, 687, 568; HRMS m/z: 350.1009 ([MþH])þ ; calcd.: 350.1022. (2E)-1-(3-Hydroxynaphthalen-2-yl)-3-(4-methoxy-3- nitrophenyl)prop-2-en-1-one 1g Mp: 112–114°C; 1 H NMR (500 MHz, CDCl3): d 12.29 (s, 1H, OH), 8.12 (dd, 1H, J ¼ 8.0, H-100 ), 8.15 (s, 1H), 8.07 (dd, 1H, J ¼ 10.5, H-50 ), 7.90 (dd, 1H, J ¼ 10.5, H-80 ), 7.79 (d, 1H, J ¼ 13.5, Hb), 7.65 (d, 1H, J ¼ 9.0, H-5), 7.62 (d, 1H, J ¼ 13.5, Ha), 7.50 (dd, 1H, J ¼ 9.0, H-60 ), 7.42 (d, 1H, J ¼ 8.5, H-6), 7.39 (m, 1H, J ¼ 7.5, H- 70 ), 7.24 (dd, 1H, J ¼ 9.5, H-30 ), 3.72 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 195.4 (C––O), 120.8 (C10 ), 162.4 (C20 ), 119.4 (C30 ), 130.2 (C40 ), 125.8 (C50 ), 130.1 (C60 ), 124.4 (C70 ), 130.4 (C80 ), 130.2 (C90 ), 142.25 (C100 ), 128.9 (vinylic), 147.5 (vinylic), 127.5 0 20 40 60 80 100 120 140 1 μM 10 μM 50 μM 100 μM 1 μM 10 μM 50 μM 100 μM control e2b2 )lortnocfo%(ytilibaivlleC Figure 5. Effect of compounds 2b and 2e on cell viability. Data are expressed as a percentage of the control. 0 50 100 150 200 250 300 control α-MSH α-MSH+1.0μM α-MSH+5.0μM α-MSH+10.0μM control α-MSH α-MSH+1.0μM α-MSH+5.0μM α-MSH+10.0μM α-MSH+kojicacid10.0μM 2e2b melanincontent(%ofcontrol) ### *** *** *** *** *** *** ***### Figure 6. Inhibitory effect of compounds 2b and 2e after treatment with 100 nM a-MSH in B16 cells. Melanin contents were measured at 405 nm. Values represent the mean Æ SE of three experiments. Data are expressed as a percentage of the control. ÃÃÃ p 0.001 compared to the group treated with 100 nm a-MSH and ### p 0.001, compared with the untreated control. 0 50 100 150 200 250 300 control α-MSH α-MSH+1.0μM α-MSH+5.0μM α-MSH+10.0μM control α-MSH α-MSH+1.0μM α-MSH+5.0μM α-MSH+10.0μM α-MSH+kojicacid10.0μM e2b2 Tyrosinaseactivity(%ofcontrol) *** *** *** *** *** *** *** ### ### Figure 7. Inhibitory effect of compounds 2b and 2e on B16 cells tyrosinase. Values represent the mean Æ SE of three experi- ments. Data are expressed as a percentage of the control. ÃÃÃ p 0.001 compared to the group treated with 100 nm a-MSH and ### p 0.001, compared with the untreated control. Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 7
  • 8. (C1), 122.5 (C2), 139.2 (C3), 147.4 (C4), 114.2 (C5), 114.2 (C6), 59.3 (Me); IR (KBr) n (cmÀ1 ): 3255, 3065, 2924, 2847, 2720, 2650, 1672, 1570, 1550, 1425, 1315, 968, 725, 580; HRMS m/z: 350.1012 ([MþH])þ ; calcd.: 350.1022. General method for the synthesis of hydroxy chalcone oxime compounds (2a–g) A mixture of chalcones 1a–g (1 mmol), hydroxylamine hydrochloride (1.5 mmol), and anhydrous sodium sulfate (1 mmol) was refluxed in ethanol (5 mL) under stirring for the period (the reaction was followed by TLC). After completion of the reaction, the mixture was filtered and the solvent was evaporated under reduced pressure. Then distilled water (10 mL) was added, and ethyl acetate (10 mL Â 3) was added to extract organic compounds. The combined organic layers were dried over anhydrous sodium sulfate and filtered. Evaporation of the solvent in reduced pressure gave the crude product, which was purified by column chromatog- raphy on silica (200–300 mesh), eluted with petroleum ether (60–90°C), or a mixture of petroleum ether and ethyl acetate (v/v ¼ 1:4) to yield the pure products (2a–g). 2-[(1E,2E)-N-Hydroxy-3-(2-methoxy-4-nitrophenyl)prop-2- enimidoyl]phenol 2a Mp: 190À192°C; 1 H NMR (500 MHz, CDCl3): d 12.52 (s, 1H, OH), 9.47 (s, 1H), 8.05 (s, 1H), 7.90 (m, 1H, J ¼ 8.5, H-6), 7.87 (d, 1H, J ¼ 12.5, Ha), 7.77 (d, 1H, J ¼ 11.0, H-60 ), 7.72 (d, 1H, J ¼ 9.5, H- 5), 7.50 (dd, 1H, J ¼ 8.0, H-40 ), 7.24 (dd, 1H, J ¼ 9.5, H-30 ), 6.94 (dd, 1H, J ¼ 10.5, H-50 ), 6.89 (d, 1H, J ¼ 12.5, Hb), 3.89 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 160.2 (C––N), 130.44 (C60 ), 120.43 (C10 ), 118.3 (C50 ), 139.2 (C40 ), 118.4 (C30 ), 164.9 (C20 ), 132.2 (C1), 114.9 (C2), 112.2 (C3), 137.5 (C4), 124.7 (C5), 118.5 (C6), 129.4 (vinylic), 142.5 (vinylic); IR (KBr) n (cmÀ1 ): 3650, 3582, 2650, 1648, 1550, 1265, 949, 835, 712; HRMS m/z: 315.0976 ([MþH])þ ; calcd.: 315.0981. (1E,2E)-N-Hydroxy-3-(2-methoxy-4-nitrophenyl)-1- (pyridin-2-yl)prop-2-en-1-imine 2b Mp: 178–180°C; 1 H NMR (500 MHz, CDCl3): d 9.42 (s, 1H), 8.77 (dd, 1H, H-3, J ¼ 10.5), 8.47 (dd, 1H, H-6, J ¼ 8.0), 8.20 (s, 1H), 7.88 (m, 1H, J ¼ 8.0, H-60 ), 7.85 (d, 1H, J ¼ 11.5, Ha), 7.80 (t,1H, H-5, J ¼ 9.5), 7.75 (d, 1H, J ¼ 9.5, H-50 ), 7.44 (dd, 1H, J ¼ 11.0, H- 4), 6.94 (d, 1H, J ¼ 11.5, Hb), 3.88 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 162.5 (C––N), 153.5 (C1), 118.9 (C6), 134.4 (C5), 130.4 (C4), 150.2 (C3), 136.4 (C10 ), 118.2 (C20 ), 114.2 (C30 ), 139.7 (C40 ), 125.3 (C50 ), 120.2 (C60 ), 144.2 (vinylic), 133.2 (vinylic); IR (KBr) n (cmÀ1 ): 3645, 3548, 2652, 1635, 1425, 1302, 1225, 805, 672; HRMS m/z: 300.0978 ([MþH])þ ; calcd.: 300.0984. 2-[(1E,2E)-N-Hydroxy-3-(4-methoxy-3-nitrophenyl)prop-2- enimidoyl]phenol 2c Mp: 165–167°C; 1 H NMR (500 MHz, CDCl3): d 12.50 (s, 1H, OH), 9.39 (s, 1H), 8.22 (s, 1H), 7.92 (d, 1H, J ¼ 11.5, Ha), 7.82 (d, 1H, J ¼ 10.0, H-60 ), 7.64 (d, 1H, J ¼ 9.0, H-5), 7.57 (d, 1H, J ¼ 8.0, H- 6), 7.44 (t, 1H, J ¼ 8.0, H-40 ), 7.35 (d, 1H, Hb, J ¼ 11.5), 7.02 (dd, 1H, J ¼ 9.5, H-50 ), 6.94 (dd, 1H, J ¼ 10.5, H-30 ), 3.92 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 160.2 (C––N), 129.9 (C60 ), 120.2 (C10 ), 118.1 (C50 ), 137.7 (C40 ), 118.2 (C30 ), 162.9 (C20 ), 124.9 (C1), 118.4 (C2), 137.2 (C3), 148.5 (C4), 114.2 (C5), 116.3 (C6), 55.8 (Me), 123.2 (vinylic), 140.9 (vinylic); IR (KBr) n (cmÀ1 ): 3659, 3545, 2650, 1625, 1400, 1312, 1234, 875, 632; HRMS m/z: 315.0975 ([MþH])þ ; calcd.: 315.0981. (1E,2E)-N-Hydroxy-3-(4-methoxy-3-nitrophenyl)-1- (pyridin-2-yl)prop-2-en-1-imine 2d Mp: 172–174°C; 1 H NMR (500 MHz, CDCl3): d 9.47 (s, 1H), 8.72 (dd, 1H, J ¼ 11.5, H-3), 8.42 (dd, 1H, J ¼ 8.5, H-6), 8.20 (s, 1H), 7.88 (m, 1H, J ¼ 8.0, H-60 ), 7.82 (t, 1H, J ¼ 9.5, H-5), 7.79 (d, 1H, J ¼ 12.5, Ha), 7.75 (d, 1H, J ¼ 9.5, H-50 ), 7.44 (dd, 1H, J ¼ 11.0, H-4), 6.92 (d, 1H, J ¼ 12.5, Hb), 3.88 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 162.5 (C––N), 153.5 (C1), 118.9 (C6), 134.4 (C5), 130.4 (C4), 150.2 (C3), 136.4 (C10 ), 118.2 (C20 ), 114.2 (C30 ), 139.7 (C40 ), 125.3 (C50 ), 120.2 (C60 ), 144.2 (vinylic), 133.2 (vinylic); IR (KBr) n (cmÀ1 ): 3645, 3548, 2652, 1635, 1425, 1302, 1225, 805, 672; HRMS m/z: 300.0976 ([MþH])þ ; calcd.: 300.0984. 2-{(1E,2E)-3-[4-(Dimethylamino)phenyl]-N-hydroxyprop-2- enimidoyl}phenol 2e Mp: 158–160°C. 1 H NMR (500 MHz CDCl3): d 12.35 (s, 1H, OH), 9.52 (s, 1H), 7.04 (dd, 2H, J ¼ 10.5, H-30 and H-50 ), 7.89 (d, 1H, J ¼ 12.5, Ha), 7.79 (d, 1H, J ¼ 7.5, H-6), 7.72 (dd, 2H, J ¼ 10.0, H- 20 and H-60 ), 7.46 (dd, 1H, J ¼ 8.0, H-4), 6.97 (t, 1H, J ¼ 9.5, H-3), 6.92 (d, 1H, J ¼ 12.5, Hb), 6.82 (dd, 1H, J ¼ 13.5, H-5); 13 C NMR (100 MHz, DMSO-d6) d 160.3 (C––N), 127.2 (C1), 131.97 (C2 and C6), 110.97 (C5 and C3), 154.33 (C4), 40.5 (N–CH3), 147.1 (vinylic), 127.2 (vinylic), 121.8 (C10 ), 160.5 (C20 ), 122.6 (C30 ), 119.7 (C40 ), 137.4 (C50 ), 134.7 (C60 ); IR (KBr) n (cmÀ1 ): 3620, 3528, 2342, 1620, 1415, 1300, 1222, 817, 632; HRMS m/z: 283.1439 ([MþH])þ ; calcd.: 283.1447. 3-[(1E,2E)-N-Hydroxy-3-(2-methoxy-4-nitrophenyl)prop-2- enimidoyl]naphthalen-2-ol 2f Mp: 145–147°C; 1 H NMR (500 MHz, CDCl3): d 12.55 (s, 1H, OH), 9.70 (s, 1H), 8.17 (s, 1H), 8.09 (dd, 1H, J ¼ 10.5, H-40 ), 7.85 (dd, 1H, J ¼ 9.0, H-80 ), 7.74 (dd, 1H, J ¼ 8.0, H-70 ), 7.58 (t, 1H, J ¼ 10.0, H-50 ), 7.57 (d, 1H, J ¼ 12.0, Hb), 7.52 (d, 1H, J ¼ 12.0, Ha), 7.35 (dd, 1H, J ¼ 8.5, H-60 ), 7.24 (dd, 1H, J ¼ 9.5, H-30 ), 7.12 (d, 1H, J ¼ 7.5, H-30 ), 3.72 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 162.5 (C––N), 126.1 (C10 ), 160.8 (C20 ), 120.1 (C30 ), 130.4 (C40 ), 126.4 (C50 ), 130.5 (C60 ), 127.2 (C70 ), 130.4 (C80 ), 129.9 (C90 ), 140.2 (C100 ), 132.2 (vinylic), 144.6 (vinylic), 58.6 (Me), 130.2 (C1), 120.8 (C2), 118.4 (C3), 129.4 (C4), 122.6 (C5), 120.2 (C6); IR (KBr) n (cmÀ1 ): 3650, 3255, 3024, 2842, 1692, 1600, 1557, 1443, 934, 720, 655, 538; HRMS m/z: 365.1131 ([MþH])þ ; calcd.: 365.1137. 3-[(1E,2E)-N-Hydroxy-3-(4-methoxy-3-nitrophenyl)prop-2- enimidoyl]naphthalen-2-ol 2g Mp: 132–134°C; 1 H NMR (500 MHz, CDCl3): d 12.05 (s, 1H, OH), 9.42 (s, 1H, OH), 8.20 (s, 1H), 7.89 (dd, 1H, J ¼ 10.5, H-5), 7.84 (dd, 1H, J ¼ 9.5, H-80 ), 7.82 (dd, 1H, J ¼ 9.5, H-6), 7.74 (dd, 1H, Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 S. K. Radhakrishnan et al. Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 8
  • 9. J ¼ 8.0, H-70 ), 7.62 (d, 1H, J ¼ 13.0, Hb), 7.52 (t, 1H, J ¼ 10.0, H- 50 ), 7.51 (dd, 1H, J ¼ 9.0, H-40 ), 7.49 (d, 1H, J ¼ 13.0, Ha), 7.24 (dd, 1H, J ¼ 9.5, H-30 ), 6.62 (dd, 1H, J ¼ 8.5, H-60 ), 3.72 (s, 3H); 13 C NMR (125 MHz, DMSO-d6) d 159.8 (C––N), 119.2 (C10 ), 120.4 (C20 ), 148.4 (C30 ), 150.2 (C40 ), 123.4 (C50 ), 128.8 (C60 ), 122.4 (C70 ), 129.4 (C80 ), 130.4 (C90 ), 140.25 (C100 ), 126.9 (vinylic), 140.5 (vinylic), 122.8 (C1), 124.5 (C2), 140.2 (C3), 140.4 (C4), 116.82 (C5), 118.2 (C6), 58.7 (Me); IR (KBr) n (cmÀ1 ): 3685, 3250, 2850, 2565, 2320, 1670, 1550, 1434, 1250, 935, 712, 535; HRMS m/z: 365.1130 ([MþH])þ ; calcd.: 365.1137. Pharmacology Tyrosinase inhibition assay The mushroom tyrosinase inhibition activity of chalcone oxime compounds was measured using L-DOPA as sub- strate [30]. Mushroom tyrosinase, L-DOPA, and tested samples were prepared by dissolving in 50 mM Na2HPO4–NaH2PO4 buffer (pH 6.8). Reaction mixtures containing 50mL of 2 mmol/L of L-DOPA, 50 mL of phosphate buffer, and 50 mL of different concentrations (0.5, 1.0, 2.5, 5.0, and 10.0 mM) of tested compounds were added in 96 well microtiter plates, followed by the addition of 50 mL of 0.2 mg/mL of mushroom tyrosinase. The assay mixture was incubated at 25°C for 30 min. Following incubation, the amount of DOPAchrome produced in the reaction mixture was determined spectro- photometrically at 492 nm using a Flex Station 3 microplate reader. We determined the inhibition at 20 mM for all the synthesized compounds. In addition, we determined the IC50, that is, the concentration of compound that causes 50% inhibition for kojic acid and the active inhibitors 2b and 2e. In the present study, dose-dependent inhibition experiments were performed in triplicate to determine the IC50 of test compounds. The average results from three experiments are shown. Determination of the inhibition type of compounds 2b and 2e on mushroom tyrosinase Inhibitors were first dissolved in DMSO and used for the test after a 30-fold dilution. The final concentration of DMSO in the test solution was 3.33%. Mushroom tyrosinase (50 mL; 0.2 mg/mL) was incubated with 50 mL of various concentrations of enzyme substrate (0.2–0.6 mM) and 50 mL of phosphate buffer, and then 50 mL of different concentrations of tested samples (0, 1.25, 5.0, and 20.0 mM) were simultaneously added to the reaction mixtures. The measurement was performed in triplicate for each concentration and averaged before further calculation. The absorbance variations from these studies were used to generate Lineweaver–Burk plots to determine the inhibition type. The kinetic parameter (Km) of the tyrosinase activity was calculated by linear regression from Lineweaver–Burk plots. For the type of enzyme inhibition and the inhibition constant (Ki) for an enzyme-inhibitor complex, the mechanisms were analyzed by Dixon plot, which is a graphical method [plot of 1/enzyme velocity (1/v) versus inhibitor concentration (I) with varying concentrations of the substrate]. In silico docking between tyrosinase and target compounds To further understand the binding modes of the most active compounds with mushroom tyrosinase, molecular docking studies of compounds 2b and 2e were performed using Discovery Studio 4.5 (Accelrys, San Diego, CA, USA). To model the tyrosinase structure, we used the crystal structure of Agaricus bisporus tyrosinase (PDB ID: 2Y9X), A chain. From the docking results, we checked for possible hydrogen-bonding and non-bonding interactions with the amino acid residues. Cell viability Cell survival was quantified by a colorimetric assay that used 3- (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and that measured mitochondrial activity in viable cells. This method is based on the conversion of MTT (Sigma) to MTT-formazan crystals by a mitochondrial enzyme. Briefly, cells seeded at a density of 3 Â 104 /cell in a 48-well plate were allowed to adhere overnight; the culture medium was then replaced with fresh serum-free DMEM. MTT was freshly prepared at 5 mg/mL in phosphate-buffered saline (PBS). Aliquots of 500 mL of MTT stock solution were added to each well, and the plate was incubated at 37°C for 4 h in a humidified 5% CO2 incubator. After 4 h, the medium was removed. To each well, 500 mL of EtOH-DMSO (solution of a 1:1 mixture) was added to dissolve the formazan. After 10 min, the optical density of each well was measured spectrophotometrically using a 560 nm filter. Assay of murine tyrosinase activity Tyrosinase activity was estimated by measuring the rate of L- DOPA oxidation. Cells were plated in 24-well dishes at a density of 5 Â 104 cells/mL. B16 cells were incubated in the presence or absence of 100 nM a-MSH, and then treated for 24 h with various concentrations of 2a or 2b (0–10 mM). Cells were washed and lysed in 100 mL of 50 mM sodium phosphate buffer (pH 6.5) containing 1% Triton X-100 (Sigma) and 0.1 mM PMSF (phenylmethylsulfonyl fluoride), and then frozen at À80°C for 30 min. After thawing and mixing, cellular extracts were clarified by centrifugation at 12000 rpm for 30 min at 4°C. An 8 mL sample of the supernatant and 20 mL of L-DOPA (2 mg/mL) were placed in a 96-well plate, and the absorbance at 492 nm was read every 10 min for 1 h at 37°C using an ELISA plate reader. The final activity is expressed as D O.D. minÀ1 for each condition. Determination of melanogenesis in B16 cells Determination of melanin content of cells was done using a modification of the method of Bilodeau et al. In the present study, the amount of melanin was used as an index of melanogenesis. B16 F10 cells (5 Â 104 ) were transferred to 24- well dishes and incubated in the presence or absence of 100 nM a-MSH. Cells were then incubated for 24 h with various concentrations of 2a and 2b (0–10 mM). After washing twice with PBS, samples were dissolved in 100 mL of 1 N NaOH. The samples were then incubated at 60°C for 1 h and mixed to Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 Novel Chalcone Oximes as Tyrosinase Inhibitors Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 9
  • 10. solubilize the melanin. Absorbance at 405 nm was compared with a standard curve of synthetic melanin. The authors have declared no conflicts of interest. References [1] R. A. Spritz, V. J. Hearing, Adv. Hum. Genet. 1994, 22, 1–45. [2] M. Brenner, V. J. Hearing, Photochem. Photobiol. 2008, 84, 539–549. [3] K. Iozumi, G. E. Hoganson, R. Pennella, M. A. Everett, B. B. Fuller, J. Invest. Dermatol. 1993, 100, 806–811. [4] H. Ando, H. Kondoh, M. Ichihashi, V. J. Hearing, J. Invest. Dermatol. 2007, 127, 751–61. [5] H. M. Wang, C. Y. Chen, Z. H. Wen, Exp. Dermatol. 2011, 20, 242–248. [6] A. B. Lerner, T. B. Fitzpatrick, E. Calkins, W. H. Summerson, J Biol Chem. 1955, 187, 793–802. [7] M. V. Martinez, J. R. Whitaker, Trends Food Sci. Tech. 1995, 6, 195–200. [8] W. Yi, R. Cao, Eur. J. Med. Chem. 2010, 45, 639–646. [9] Y.Xu,A.H.Stokes,W.M.Freeman,S.C. Kumer, B. A.Vogt, K. E. Vrana, Mol. Brain Res. 1997, 45(1), 159–162. [10] M. Ashida, P. Brey, Proc. Natl. Acad. Sci. USA 1995, 92, 10698–10702. [11] S. Passi, P. M. Nazzaro, Br. J. Dermatol. 1981, 104, 659–665. [12] M. Criton, V. Mellay-Hamon, Bioorg. Med. Chem. Lett. 2008, 18, 3607–3610. [13] H. S. Baek, H. S. Rho, J. W. Yoo, S. M. Ahn, J. Y. Lee, J. Lee, M. K. Kim, D. K. Kim, I. S. Chang, Bull. Kor. Chem. Soc. 2008, 29, 43–46. [14] H. J. Lindner, S. Goettlicher, Acta Crystallogr. Sect. B 1969, 25, 832–842. [15] S. K. Radhakrishnan, R. Shimmon Conn, A. T. Baker, Bioorg. Med. Chem. Lett. 2015, 25, 1753–1756. [16] C. Q. Meng, L. Ni, K. J. Worsencroft, Z. Ye, M. D. Weingarten, J. E. Simpson, J. W. Skudlarek, E. M. Marino, K. L. Suen, C. Kunsch, A. Souder, R. B. Howard, C. L. Sundell, M. A. Wasserman, J. A. Sikorski, J. Med. Chem. 2007, 50, 1304–1315. [17] T. W. Yan, J. Q. Ya, L. Z. Ya, J. L. Yu, R. Bing, Q. Z. Yan, R. Y. Meng, Q. J. Ai, L. Q. Jin, L. Z. Hai, RSC Adv. 2014, 61, 32263–32275. [18] Y. Matoba, T. Kumagai, A. Yamamoto, H. Yoshitsu, M. Sugiyama, J. Biol. Chem. 2008, 281, 8981–8990. [19] H. Decker, F. Tuczek, Trends Biochem. Sci. 2000, 25, 392–397. [20] M. Jimenez-Atienzar, J. Escribano, J. Cabanes, F. Gandıa- Herrero, F. Garcıa-Carmona, Plant Physiol. Biochem. 2005, 43, 866–873. [21] S. Ennes, R. Paschoalick, M. M. D. A. Alchorne, J. Dermatol. Treat. 2000, 11, 173–179. [22] P. G. Engasser, J. Am. Acad. Dermatol. 1984, 10, 1072–1073. [23] A. Fisher, Cutis 1983, 31, 240–244, 250. [24] C. Romaguera, F. Grimalt, Contact Dermatitis 1985, 12, 183. [25] E. V. Curto, C. Kwong, H. Hermersdorfer, H. Glatt, C. Santis, V. Virador, V. J. Hearing, T. P. Dooley, Biochem. Pharmacol. 1999, 57, 663–672. [26] H. Zhou, J. K. Kepa, D. Siegel, S. Miura, Y. Hiraki, D. Ross, Mol. Pharmacol. 2009, 76, 579–587. [27] V. Kahn, Pigment Cell Res. 1995, 8, 234–240. [28] N. Fujimoto, H. Onodera, K. Mitsumori, T. Tamura, S. Maruyama, A. Ito, Carcinogenesis 1999, 20, 1567–1571. [29] Y. J. Zhu, L. Qiu, J. J. Zhou, H. Y. Guo, Y. H. Hu, Z. C. Li, Q. Wang, Q. X. Chen, B. Liu, J. Enz. Inh. Med. Chem. 2010, 25, 798–803. [30] J. K. No, D. Y. Soung, Y. J. Kim, K. H. Shim, Y. S. Jun, S. H. Rhee, T. Yokozawa, H. Y. Chung, Life Sci. 1999, 65, 241–246. Arch. Pharm. Chem. Life Sci. 2015, 348, 1–10 S. K. Radhakrishnan et al. Archiv der Pharmazie ARCHRCH PHARMHARM ß 2015 WILEY-VCH Verlag GmbH Co. KGaA, Weinheim www.archpharm.com 10