SlideShare une entreprise Scribd logo
1  sur  5
Télécharger pour lire hors ligne
Inhibitory kinetics of novel 2,3-dihydro-1H-inden-1-one
chalcone-like derivatives on mushroom tyrosinase
Sini Radhakrishnan ⇑
, Ronald Shimmon, Costa Conn, Anthony Baker
School of Chemistry and Forensic Science, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
a r t i c l e i n f o
Article history:
Received 1 August 2015
Revised 20 October 2015
Accepted 23 October 2015
Available online xxxx
Keywords:
Tyrosinase
Competitive
Diphenolase
Reversible
a b s t r a c t
In this study, novel 2,3-dihydro-1H-inden-1-one chalcone-like compounds and their hydroxyl derivatives
were synthesized, and their inhibitory effects on mushroom tyrosinase activity were evaluated. Two of
the compounds synthesized inhibited the diphenolase activity of tyrosinase in a dose dependent manner
and exhibited much higher tyrosinase inhibitory activities (IC50 values of 12.3 lM and 8.2 lM, respec-
tively) than the positive control, kojic acid (IC50: 27.5 lM). Kinetic analysis showed that their inhibition
was reversible. Both the novel compounds displayed competitive inhibition with their Ki values of
10.3 lM and 8.7 lM, respectively. This study suggests hydroxy substituted 2,3-dihydro-1H-inden-1-
one chalcone-like compounds to serve as promising candidates for use as depigmentation agents.
Ó 2015 Elsevier Ltd. All rights reserved.
Melanin, a polymeric dark pigment is produced by melanocytes
within specialized organelles called melanosomes. The melano-
somes contain several enzymes that mediate the production of
melanin.1,2
The conversion of tyrosine to melanin requires
tyrosinase, also known as polyphenol oxidase, a copper containing
protein. Tyrosinase [EC 1.14.18.1], is the key enzyme in the mela-
nogenic pathway responsible for the hydroxylation of L-tyrosine
to 3,4-dihydroxy phenylalanine (L-DOPA) and oxidation of L-DOPA
to dopaquinone.3,4
However, an excessive accumulation of the
pigment could lead to serious aesthetic issues. Alterations in
tyrosinase function could culminate with serious dermatological
http://dx.doi.org/10.1016/j.bmcl.2015.10.071
0960-894X/Ó 2015 Elsevier Ltd. All rights reserved.
⇑ Corresponding author. Tel.: +61 426687300.
E-mail address: Sini.KaranayilRadhakrishnan@student.uts.edu.au
(S. Radhakrishnan).
O
+ O
R
1
H
a
O
R
1
Scheme 1. General method for synthesis of 2,3-dihydro-1H-inden-1-one chalcones.
Reagents and conditions: (a) MeOH, NaOH, 0 °C, 24 h.
Table 1
Substitution pattern and tyrosinase inhibition effects of 1-indanone derivatives
O
R
1
R
2
R
3
R
4
Compound R1
R2
R3
R4
Yield
(%)
Inhibition rate (%) at
50 lM
1a H NO2 H H 95.5 22.6 ± 0.44
1b H H NO2 H 87.6 19.8 ± 0.15
1c OMe H NO2 H 64.2 25.4 ± 0.55
1d H NO2 OMe H 54.1 24.2 ± 0.33
1e OMe H OMe H 85.4 30.6 ± 0.47
1f H OMe OMe H 77.2 32.2 ± 0.11
1g H OMe H OMe 70.3 38.6 ± 0.32
2a OH H OH H 51.2 65.2 ± 0.15
2b H OH OH H 45.2 74.6 ± 0.14
2c H OH H OH 42.6 50.1 ± 0.38
Kojic acid 45.2 ± 0.22
O
OMe b
O
OH
Scheme 2. Dealkylation of methoxy chalcones. Reagents and conditions: (b) BBr3,
CH2Cl2.
Bioorganic & Medicinal Chemistry Letters xxx (2015) xxx–xxx
Contents lists available at ScienceDirect
Bioorganic & Medicinal Chemistry Letters
journal homepage: www.elsevier.com/locate/bmcl
Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
disorders including melasma, ephelides (freckles), solar lentigo
(age spots), and sites of actinic damage.5–7
Tyrosinase is responsi-
ble for melanization in animals, cuticle formation in insects,
wound healing and browning of fruits and vegetables.8–15
There-
fore, tyrosinase inhibitors have potential applications in the treat-
ment of dermatological disorders associated with melanin
hyperpigmentation, in agriculture as bio-insecticides and also in
cosmetics for whitening and depigmentation after sunburn. From
a structural perspective, tyrosinase has two copper ions in its
active site which play a vital role in its catalytic activity. At the
active site of tyrosinase, a dioxygen molecule binds in side-on
coordination between two copper ions. Each of the copper ions is
coordinated by three histidines in the protein matrix. 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 produc-
tion of the brown color.16,17
Several natural and synthetic tyrosinase inhibitors have been
reported, including aromatic aldehydes and acids, tropolone,
arbutin, flavonoids, and kojic acid.18,19
However, most of these
compounds have been reported to be either toxic towards cells
or have low stability towards oxygen and water.20–22
Indanones are bioactive molecules that are frequently used as
precursors in the synthesis of pharmaceutical and natural product
substances. Previously, indanone based curcumin analogs have
been reported to have strong inhibitory effects on tyrosinase.23
Another indanone derivative, tetrahydrocurcumin, was recom-
mended to be used in cosmetics as a lighting agent.24
Although indole and its derivatives are popular medicinal
agents, the related heterocycles like indene and their derivatives
like substituted 1-indanones (2,3-dihydro-1H-inden-1-one) have
much less been exploited for their biological potential. Previously
we have studied the tyrosinase inhibition of azachalcone com-
pounds.25
Hence, we considered that it might be interesting to syn-
thesize a series of novel 2,3-dihydro-1H-inden-1-one chalcone-like
0
20
40
60
80
100
120
0 5 10 15 20
Relativeactivity(%)
[I] (µM)
2a
2b
Figure 1. Inhibition effects of compounds 2a and 2b on the diphenolase activity of
mushroom tyrosinase. Data are presented as means (n = 3).
0
50
100
150
200
250
0 2 4 6 8 10 12
Activity(µM/min)
[E] (µg/mL)
0
1
2
3
4
0
20
40
60
80
100
120
140
160
180
0 2 4 6 8 10 12
Activity(µM/min)
[E] (µg/mL)
4
1
2
3
0
2b
2a
Figure 2. The inhibitory mechanism of compounds 2a and 2b on mushroom tyrosinase were reversible. The concentration of inhibitor used for curves 0–4 are 0, 0.25, 0.5, 1.0
and 2.0 lM, respectively.
2 S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx
Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
compounds and their hydroxyl derivatives for use as depigmenta-
tion agents and as anti-browning food additives.
In the first step, 2,3-dihydro-1H-inden-1-one chalcone-like
compounds were synthesized by the base catalyzed Claisen–
Schmidt condensation of 1-indanone and an appropriate ketone
in a polar solvent like methanol (Scheme 1). Some selected
methoxy chalcones were then successfully dealkylated to their
corresponding hydroxy compounds in the presence of boron
tribromide (BBR3) (Scheme 2).
In the present study, tyrosinase extracted from the edible
mushroom Agaricus bisporus is used due to its easy availability
and high homology with the mammalian enzyme that renders it
well suited as a model for studies on melanogenesis.26
We used
L-DOPA as substrate to study the effect of inhibitor compounds
on the oxidation of L-DOPA by tyrosinase (diphenolase activity).27
Kojic acid, a well-known tyrosinase inhibitor served as the positive
control. We have further investigated the kinetic parameters and
inhibition mechanisms of active tyrosinase inhibitor compounds.
Dose-dependent inhibition experiments were performed in tripli-
cate to determine the IC50 of the active inhibitors 2a and 2b.
In terms of the structure–activity relationships, compound 2b
((2Z)-2-(3,4-dihydroxybenzylidene)-2,3-dihydro-1H-inden-1-one)
exhibited the most potent tyrosinase inhibitory activity with
inhibition of 74.6%. This could be accounted to the presence of a
3,4-dihydroxy group in the B-ring that shows structural
resemblance with the substrate, L-DOPA. Compound 2a with an
ortho–para dihydroxy substitution on ring B showed an inhibition
of 65.2% at 50 lM. The inhibition of both compounds 2a and 2b
was seen to be greater than the positive control kojic acid which
exhibited 45.2% inhibition at 50 lM. The inhibitory activities of
these newly synthesized compounds were compared to that of
kojic acid as a reference standard and the results are shown in
Table 1.
Results indicated that both compounds 2a and 2b could inhibit
the diphenolase activity of tyrosinase in a dose-dependent manner.
With increasing concentrations of inhibitors, the remaining
enzyme activity decreased exponentially. The IC50 for compounds
2a and 2b was estimated to be 12.3 lM and 8.2 lM, respectively
(Fig. 1).
The plots of the remaining enzyme activity versus the concen-
tration of enzyme at different inhibitor concentrations gave a
family of straight lines, which all passed through the origin. The
presence of inhibitor did not reduce the amount of enzyme, but
just resulted in the inhibition of enzyme activity. The results
showed that both the compounds 2a and 2b were reversible inhi-
bitors of mushroom tyrosinase for oxidation of L-DOPA (Fig. 2).
Compounds 1c, 2b, 2c and 2d exhibited moderate tyrosinase
inhibition while compounds 1a, 1b and 1d–1f showed negligible
tyrosinase inhibition in comparison with kojic acid. Previous stud-
ies have recognized potent inhibitory effect of chalcones with
hydroxyl groups on tyrosinase.28–34
The hydroxyl groups in com-
pounds carry out the nucleophilic attack on the coppers of tyrosi-
nase active site and directly involved in transferring protons during
catalysis, which resulted in inactivation of tyrosinase.35,36
Further-
more, results indicated that electron-donating groups contributed
more to the inhibitory activity of the compounds on mushroom
-200
-100
0
100
200
300
400
-4 -2 0 2 4 6
1/V(µM/min)-1
1/S (mM/L)
2a
-200
-100
0
100
200
300
400
-4 -2 0 2 4 6
1/V(µM/min)-1
1/S (mM/L)
2b
Figure 3. Lineweaver Burk plot for inhibition of compounds 2a and 2b on mushroom tyrosinase. Data were obtained as mean values of 1/V, the inverse of the absorbance
increase at a wavelength of 492 nm per min of three independent tests with different concentrations of L-DOPA as a substrate. The concentration of compounds 2a and 2b
from top to bottom is 20 lM, 5 lM, 1.25 lM and 0 lM, respectively.
S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx 3
Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
tyrosinase than the electron-withdrawing groups. Electron donat-
ing groups (e.g., –OMe, –OH) on the atoms adjacent to the p system
activate the aromatic ring by increasing the electron density on the
ring through a resonance donating effect.
In this study, we investigated in depth the tyrosinase inhibitory
activities of compounds 2b and 2a. We measured the reaction rates
in the presence of active inhibitors with various concentrations of
L-DOPA as a substrate. As the concentrations of active inhibitors 2a
and 2b increased, Km values gradually increased, but Vmax values
did not change, thereby indicating the inhibitors act as competitive
inhibitors of mushroom tyrosinase (Fig. 3).
The inhibition kinetics were illustrated by Dixon plots, which
were obtained by plotting 1/V versus [I] with varying concentra-
tions of substrate. Dixon plots gave a family of straight lines pass-
ing through the same point at the second quadrant, giving the
inhibition constant (Ki) (Fig. 4).
The Ki value for the compounds 2a and 2b estimated from this
Dixon plot was 10.3 lM and 8.7 lM, respectively. A comparison of
the Ki values of the compounds with that of kojic acid revealed that
they possess much higher affinity to tyrosinase than kojic acid
(Table 2).
To summarize, in this study, we synthesized novel 2,3-dihydro-
1H-inden-1-one (1-indanone) chalcone-like compounds and
their hydroxy derivatives, and studied their inhibition on the
diphenolase activity of mushroom tyrosinase. Hydroxy substituted
1-indanone chalcone-like compounds 2a and 2b were found to be
significantly more potent than kojic acid with their IC50 values of
12.3 lM and 8.2 lM, respectively. Both the compounds exhibited
reversible competitive inhibition. Compound 2b with a 3,4-dihy-
droxy group in the B-ring had structural resemblance to the
substrate L-DOPA. This leads to the competitive displacement of
L-DOPA from the active site of the enzyme tyrosinase in a lock-
and key model. In addition, the presence of the hydroxy group at
the 4-position (ring B) is indispensable for significant tyrosinase
inhibition in this class of compounds. These preliminary findings
justify pursuing further work on hydroxy substituted 1-indanone
chalcone-like compounds to serve as a potential scaffold for the
future development of active tyrosinase inhibitors.
Notes: 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
-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
-30
-20
-10
0
10
20
30
40
-40 -30 -20 -10 0 10 20 30
1/V(µM/min)-1
[inhibitor] µM
2a
Figure 4. Dixon plot for the inhibitory effect of compounds 2a and 2b on L-DOPA oxidation catalyzed by mushroom tyrosinase. The inhibitor concentrations were 0, 10 lM
and 20 lM, respectively. The L-DOPA concentrations were 200, 400 and 600 lM.
Table 2
Effect on mushroom polyphenol oxidase activity and kinetic analysis of compounds
Compound Type of inhibition IC50 (lM) RA*
Ki
#
(lM)
2a Competitive 12.3 ± 0.22 2.23F 10.3
2b Competitive 8.2 ± 0.44 3.35F 8.7
Kojic acid Competitive 27.5 ± 0.56 12.2
#
Values were measured at 5 lM of active compounds and Ki is the (inhibitor
constant).
*
RA is the relative inhibitory activity compared to the standard kojic acid where
1.0F means one time activity of kojic acid.
4 S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx
Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
chemical shifts (o) are quoted in parts per million downfield from
TMS and coupling constants (J) are given in Hz. Abbreviations used
in the splitting pattern were as follows: s = singlet, d = doublet,
t = triplet, m = multiplet. 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
purchased from Sigma–Aldrich Chemical Co.
General method for the synthesis of 2,3-dihydro-1H-inden-1-one
(1-indanone) chalcone-like compounds (1a–1g): To a stirred solution
of 1-indanone (2.0 mmol) and the corresponding aldehyde
(2.0 mmol) in methanol (6 mL) at room temperature was added
freshly pulverized sodium hydroxide (160 mg, 4.0 mmol). The
solution was stirred overnight (monitored by TLC) and then poured
onto crushed ice. Once all the ice had melted, pH was adjusted to 5
using hydrochloric acid (5 M). The resulting solution was then
extracted with ethyl acetate, washed with brine and filtered after
drying with anhydrous sodium sulfate. The solution was then
concentrated with a rotary evaporator under vacuum to give the
respective compound.
Method for the synthesis of hydroxy indanone compounds (2a–2c):
A solution of BBr3 (2.5 mL for each methoxy group) was added to a
cooled solution of the corresponding methoxy chalcone (1 mmol)
in CH2Cl2 under argon. The cooling bath was removed and the dark
solution was warmed to rt and stirred for 1–5 h. The dark solution
was then poured into ice water and filtered. The aqueous layer was
extracted with chloroform twice. The organic extract was washed
with water followed by brine and anhydrous sodium sulfate. This
was then rotary evaporated to give the respective product.
Enzyme activity assay: The mushroom tyrosinase inhibition
activity of 1-indanone chalcone-like compounds was measured
using L-DOPA as substrate. The activity assay used 3 ml of reaction
medium containing 0.5 mM L-DOPA in 50 mM Na2HPO4–NaH2PO4
buffer (pH 6.8). The final concentrations of tyrosinase was
3.33 mg/ml. The substrate reaction progress curve was analyzed
to obtain the reaction rate constants. The reaction was carried
out at 30 °C and pH 6.8.
Determination of the inhibition type of compounds 2a and 2b 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 lL; 0.2 mg mLÀ1
) was incubated with 50 lL of various concen-
trations of enzyme substrate and 50 lL of phosphate buffer, and
then 50 lL of different concentrations of tested samples 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 ana-
lyzed by Dixon plot, which is a graphical method plot of 1/enzyme
velocity (1/V) versus inhibitor concentration (I) with varying con-
centrations of the substrate.
Supplementary data
Supplementary data associated with this article can be found, in
the online version, at http://dx.doi.org/10.1016/j.bmcl.2015.10.
071.
References and notes
1. Hearing, V. J. Methods Enzymol. 1987, 142, 154.
2. Spritz, R. A.; Hearing, V. J. Adv. Hum. Genet. 1994, 22, 1.
3. Cooksey, C. J.; Garratt, P. J.; Land, E. J.; Pavel, S.; Ramsden, C. A.; Riley, P. A.;
Smit, N. P. J. Biol. Chem. 1997, 272, 26226.
4. Kubo, I.; Kinst-Hori, I.; Chaudhuri, S. K.; Kubo, Y.; Ogura, Y. T. Bioorg. Med. Chem.
2000, 8, 1749.
5. Iozumi, K.; Hoganson, G. E.; Pennella, R.; Everett, M. A.; Fuller, B. B. J. Invest.
Dermatol. 1993, 100, 806.
6. Ando, H.; Kondoh, H.; Ichihashi, M.; Hearing, V. J. Invest. Dermatol. 2007, 127,
751.
7. Wang, H. M.; Chen, C. Y.; Wen, Z. H. Exp. Dermatol. 2011, 20, 242.
8. Olivares, C.; Solano, F. Pigment Cell Melanoma Res. 2009, 22, 750.
9. Jimbow, K.; Park, J. S.; Kato, F.; Hirosaki, K.; Toyofuku, K.; Hua, C.; Yamashita, T.
Pigment Cell Res. 2000, 13, 222.
10. Kim, Y. J.; Uyama, H. Cell. Mol. Life Sci. 2005, 62, 1707.
11. Rescigno, A.; Sollai, F.; Pisu, B.; Rinaldi, A.; Sanjust, E. J. Enzyme Inhib. Med.
Chem. 2002, 17, 207.
12. Kanost, M. R.; Jiang, H.; Yu, X. Q. Immunol. Rev. 2004, 198, 97.
13. Lai, S. C.; Chen, C. C.; Hou, R. R. J. Med. Entomol. 2002, 39, 266.
14. Guerrero, A.; Rosell, G. Curr. Med. Chem. 2005, 12, 461.
15. Gorman, M. J.; An, C.; Kanost, M. R. Insect Biochem. Mol. Biol. 2007, 37, 1327.
16. Sanchez-Ferrer, A.; Rodriguez-Lopez, J. N.; Garcia-Canavas, F.; Garcia-Carmona,
F. Biochim. Biophys. Acta 1995, 1, 1247.
17. Decker, H.; Schweikardt, T.; Nillius, D.; Salzbrunn, U.; Jaenicke, E.; Tuczek, F.
Gene 2007, 398, 183.
18. Chen, Q. X.; Song, K. K.; Qiu, L.; Liu, X. D.; Huang, H.; Guo, H. Y. Food Chem. 2005,
91, 269.
19. Xie, L. P.; Chen, Q. X.; Huang, H.; Wang, H. Z.; Zhang, R. Q. Biochemistry 2003, 68,
487.
20. Burnett, C. L.; Bergfeld, W. F.; Belsito, D. V.; Hill, R. A.; Klaassen, C. D.; Liebler, D.
C.; Marks, J. G.; Shank, R. C.; Slaga, T. J.; Snyder, P. W.; Andersen, F. A. Int. J.
Toxicol. 2010, 29, 244.
21. Draelos, Z. D. Dermatol. Ther. 2007, 20, 308.
22. Loizzo, M. R.; Tundiz, T.; Menichini, F. Compr. Rev. Food. Sci. F 2012, 11, 378.
23. Sugimoto, H. Pure Appl. Chem. 1999, 71, 2031.
24. Asawanonda, P.; Klahan, S. O. Photomed. Laser Surg. 2010, 28, 679.
25. Radhakrishnan, K. S.; Shimmon, R.; Conn, C.; Baker, T. A. Bioorg. Med. Chem. Lett.
2015, 25, 1753.
26. Song, K. K.; Chen, Q. X.; Wang, Q.; Qiu, L.; Huang, H. J. Enzyme Inhib. Med. Chem.
2005, 20, 239.
27. Gerdemann, C.; Eicken, C.; Krebs, B. Acc. Chem. Res. 2002, 35, 183.
28. Yan, Q.; Cao, R.; Yi, W.; Yu, L.; Chen, Z.; Ma, L.; Song, H. Bioorg. Med. Chem. Lett.
2009, 19, 4055.
29. Yamazaki, Y.; Kawano, Y.; Yamanaka, A.; Maruyama, S. Bioorg. Med. Chem. Lett.
2009, 19, 4178.
30. Kanade, S. R.; Suhas, V. L.; Chandra, N.; Gowda, L. R. FEBS J. 2007, 274, 4177.
31. Kim, D.; Park, J.; Kim, J.; Han, C.; Yoon, J.; Kim, N.; Seo, J.; Lee, C. J. Agric. Food
Chem. 2006, 54, 935.
32. Shiino, M.; Watanabe, Y.; Umezawa, K. Bioorg. Chem. 2003, 31, 129.
33. Shiino, M.; Watanabe, Y.; Umezawa, K. Bioorg. Med. Chem. 2001, 9, 1233.
34. Yokota, T.; Nishio, H.; Kubota, Y.; Mizoguchi, M. Pigment Cell Res. 1998, 11, 355.
35. Mu~noz-Mu~noz, J. L.; Garcia-Molina, F.; Garcia-Ruiz, P. A.; Molina-Alarcon,
M.; Tudela, J.; Garcia-Canovas, F.; Rodriguez-Lopez, J. N. Biochem. J. 2008, 416,
431.
36. Espin, J. C.; Varon, R.; Fenoll, L. G.; Gilabert, M. A.; Garcia-Ruiz, P. A.; Tudela, J.;
Garcia-Canovas, F. Eur. J. Biochem. 2000, 267, 1270.
S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx 5
Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071

Contenu connexe

Tendances

DPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal Plants
DPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal PlantsDPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal Plants
DPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal Plants
IOSR Journals
 
Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...
Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...
Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...
GC University Faisalabad
 
Study of invitro anti-Oxidant Activity of Ipomea Pes-Caprae
Study of invitro anti-Oxidant Activity of Ipomea Pes-CapraeStudy of invitro anti-Oxidant Activity of Ipomea Pes-Caprae
Study of invitro anti-Oxidant Activity of Ipomea Pes-Caprae
SriramNagarajan19
 

Tendances (20)

TIME EFFICIENT BAYLIS-HILLMAN REACTION ON STEROIDAL NUCLEUS OF WITHAFERIN-A T...
TIME EFFICIENT BAYLIS-HILLMAN REACTION ON STEROIDAL NUCLEUS OF WITHAFERIN-A T...TIME EFFICIENT BAYLIS-HILLMAN REACTION ON STEROIDAL NUCLEUS OF WITHAFERIN-A T...
TIME EFFICIENT BAYLIS-HILLMAN REACTION ON STEROIDAL NUCLEUS OF WITHAFERIN-A T...
 
DPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal Plants
DPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal PlantsDPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal Plants
DPPH Scavenging Assay of Eighty Four Bangladeshi Medicinal Plants
 
Antioxidant analysis
Antioxidant analysisAntioxidant analysis
Antioxidant analysis
 
2011 acetylcholinesterase inhibiting alkaloids from
2011 acetylcholinesterase inhibiting alkaloids from2011 acetylcholinesterase inhibiting alkaloids from
2011 acetylcholinesterase inhibiting alkaloids from
 
paper1-published
paper1-publishedpaper1-published
paper1-published
 
Hypervalent organo iodines reagents in organic synthesis
Hypervalent  organo iodines reagents in organic synthesisHypervalent  organo iodines reagents in organic synthesis
Hypervalent organo iodines reagents in organic synthesis
 
Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...
Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...
Bioactive compounds and antioxidant capacities of fresh and canned fruit,of p...
 
Ntioxidant study of usnic acid and its derivative usnic acid diacetate
Ntioxidant study of usnic acid and its derivative usnic acid diacetateNtioxidant study of usnic acid and its derivative usnic acid diacetate
Ntioxidant study of usnic acid and its derivative usnic acid diacetate
 
A review on antioxidant methods
A review on antioxidant methodsA review on antioxidant methods
A review on antioxidant methods
 
paper 2 published
paper 2 publishedpaper 2 published
paper 2 published
 
Anti Bacterial and Anti Oxidant Activities of Evolvulus Alsinoides Linn
Anti Bacterial and Anti Oxidant Activities of Evolvulus Alsinoides LinnAnti Bacterial and Anti Oxidant Activities of Evolvulus Alsinoides Linn
Anti Bacterial and Anti Oxidant Activities of Evolvulus Alsinoides Linn
 
General Laboratory Assays
General Laboratory AssaysGeneral Laboratory Assays
General Laboratory Assays
 
How to find antioxidant properties from selected plants
How to find antioxidant properties from selected plantsHow to find antioxidant properties from selected plants
How to find antioxidant properties from selected plants
 
Masud
MasudMasud
Masud
 
Study of invitro anti-Oxidant Activity of Ipomea Pes-Caprae
Study of invitro anti-Oxidant Activity of Ipomea Pes-CapraeStudy of invitro anti-Oxidant Activity of Ipomea Pes-Caprae
Study of invitro anti-Oxidant Activity of Ipomea Pes-Caprae
 
A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...
A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...
A Review: 3, 4-Dihydropyrimidines Thione Their Chemistry and Pharmacological ...
 
International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)
 
Comparative Antioxidant study of gall induced plant leaves
Comparative Antioxidant study of gall induced plant leavesComparative Antioxidant study of gall induced plant leaves
Comparative Antioxidant study of gall induced plant leaves
 
acs.biomac.5b00657
acs.biomac.5b00657acs.biomac.5b00657
acs.biomac.5b00657
 
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
 

En vedette (11)

Data Memorável
Data MemorávelData Memorável
Data Memorável
 
Juguetes responsables, juguetes intercambiables
Juguetes responsables, juguetes intercambiablesJuguetes responsables, juguetes intercambiables
Juguetes responsables, juguetes intercambiables
 
OracióNam..
OracióNam..OracióNam..
OracióNam..
 
Psicoprensa
PsicoprensaPsicoprensa
Psicoprensa
 
13-09-2009 Muestras de apoyo por parte del público recibió el Gobernador a s...
13-09-2009  Muestras de apoyo por parte del público recibió el Gobernador a s...13-09-2009  Muestras de apoyo por parte del público recibió el Gobernador a s...
13-09-2009 Muestras de apoyo por parte del público recibió el Gobernador a s...
 
Verbs
VerbsVerbs
Verbs
 
600.33 transfer of ownership
600.33 transfer of ownership600.33 transfer of ownership
600.33 transfer of ownership
 
Ppt17028
Ppt17028Ppt17028
Ppt17028
 
Wood Turning 1
Wood Turning 1Wood Turning 1
Wood Turning 1
 
開発者のためのiPhone⇔Androidアプリ移植のポイント
開発者のためのiPhone⇔Androidアプリ移植のポイント開発者のためのiPhone⇔Androidアプリ移植のポイント
開発者のためのiPhone⇔Androidアプリ移植のポイント
 
環保署完成資源循環利用法(草案),邁向資源循環零廢棄之願景
環保署完成資源循環利用法(草案),邁向資源循環零廢棄之願景環保署完成資源循環利用法(草案),邁向資源循環零廢棄之願景
環保署完成資源循環利用法(草案),邁向資源循環零廢棄之願景
 

Similaire à paper 5

PBL 490 Publication
PBL 490 PublicationPBL 490 Publication
PBL 490 Publication
Jen Klabis
 
11.synthesis, characterization, antimicrobial
11.synthesis, characterization, antimicrobial11.synthesis, characterization, antimicrobial
11.synthesis, characterization, antimicrobial
Alexander Decker
 
Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...
Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...
Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...
iosrjce
 

Similaire à paper 5 (20)

paper 6 published
paper 6 publishedpaper 6 published
paper 6 published
 
paper 7 published
paper 7 publishedpaper 7 published
paper 7 published
 
Paper 3
Paper 3Paper 3
Paper 3
 
PBL 490 Publication
PBL 490 PublicationPBL 490 Publication
PBL 490 Publication
 
bio org chem
bio org chembio org chem
bio org chem
 
11.synthesis, characterization, antimicrobial
11.synthesis, characterization, antimicrobial11.synthesis, characterization, antimicrobial
11.synthesis, characterization, antimicrobial
 
Synthesis, characterization, antimicrobial
Synthesis, characterization, antimicrobialSynthesis, characterization, antimicrobial
Synthesis, characterization, antimicrobial
 
STUDIES ON TREATMENT OF PHARMACEUTICAL WASTE EFFLUENTS BY POLYMER MATERIALS M...
STUDIES ON TREATMENT OF PHARMACEUTICAL WASTE EFFLUENTS BY POLYMER MATERIALS M...STUDIES ON TREATMENT OF PHARMACEUTICAL WASTE EFFLUENTS BY POLYMER MATERIALS M...
STUDIES ON TREATMENT OF PHARMACEUTICAL WASTE EFFLUENTS BY POLYMER MATERIALS M...
 
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
 
Extraction, identification and antioxidant activities of carotenoids from Ipo...
Extraction, identification and antioxidant activities of carotenoids from Ipo...Extraction, identification and antioxidant activities of carotenoids from Ipo...
Extraction, identification and antioxidant activities of carotenoids from Ipo...
 
Synthesis, Characterization and Biological Evaluation of Oxazolone Derivatives
Synthesis, Characterization and Biological Evaluation of Oxazolone DerivativesSynthesis, Characterization and Biological Evaluation of Oxazolone Derivatives
Synthesis, Characterization and Biological Evaluation of Oxazolone Derivatives
 
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complexElectrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
 
JSEHR 1(1)-4
JSEHR 1(1)-4JSEHR 1(1)-4
JSEHR 1(1)-4
 
Molecules 22-00357
Molecules 22-00357Molecules 22-00357
Molecules 22-00357
 
Photodegradation of Hexythiaz ox in Different Solvent Systems under the Influ...
Photodegradation of Hexythiaz ox in Different Solvent Systems under the Influ...Photodegradation of Hexythiaz ox in Different Solvent Systems under the Influ...
Photodegradation of Hexythiaz ox in Different Solvent Systems under the Influ...
 
Ameliorating Effect of Frankincense on Red Blood Cells of Alloxan Induced-Dia...
Ameliorating Effect of Frankincense on Red Blood Cells of Alloxan Induced-Dia...Ameliorating Effect of Frankincense on Red Blood Cells of Alloxan Induced-Dia...
Ameliorating Effect of Frankincense on Red Blood Cells of Alloxan Induced-Dia...
 
Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...
Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...
Design, synthesis, characterization and biological evaluation of 3- (4-(7-chl...
 
Toxic Effect of Glyphosate-Pesticide on Lipid Peroxidation Superoxide Dismuta...
Toxic Effect of Glyphosate-Pesticide on Lipid Peroxidation Superoxide Dismuta...Toxic Effect of Glyphosate-Pesticide on Lipid Peroxidation Superoxide Dismuta...
Toxic Effect of Glyphosate-Pesticide on Lipid Peroxidation Superoxide Dismuta...
 
5. C.J. Patil Article.pdf
5. C.J. Patil Article.pdf5. C.J. Patil Article.pdf
5. C.J. Patil Article.pdf
 

paper 5

  • 1. Inhibitory kinetics of novel 2,3-dihydro-1H-inden-1-one chalcone-like derivatives on mushroom tyrosinase Sini Radhakrishnan ⇑ , Ronald Shimmon, Costa Conn, Anthony Baker School of Chemistry and Forensic Science, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia a r t i c l e i n f o Article history: Received 1 August 2015 Revised 20 October 2015 Accepted 23 October 2015 Available online xxxx Keywords: Tyrosinase Competitive Diphenolase Reversible a b s t r a c t In this study, novel 2,3-dihydro-1H-inden-1-one chalcone-like compounds and their hydroxyl derivatives were synthesized, and their inhibitory effects on mushroom tyrosinase activity were evaluated. Two of the compounds synthesized inhibited the diphenolase activity of tyrosinase in a dose dependent manner and exhibited much higher tyrosinase inhibitory activities (IC50 values of 12.3 lM and 8.2 lM, respec- tively) than the positive control, kojic acid (IC50: 27.5 lM). Kinetic analysis showed that their inhibition was reversible. Both the novel compounds displayed competitive inhibition with their Ki values of 10.3 lM and 8.7 lM, respectively. This study suggests hydroxy substituted 2,3-dihydro-1H-inden-1- one chalcone-like compounds to serve as promising candidates for use as depigmentation agents. Ó 2015 Elsevier Ltd. All rights reserved. Melanin, a polymeric dark pigment is produced by melanocytes within specialized organelles called melanosomes. The melano- somes contain several enzymes that mediate the production of melanin.1,2 The conversion of tyrosine to melanin requires tyrosinase, also known as polyphenol oxidase, a copper containing protein. Tyrosinase [EC 1.14.18.1], is the key enzyme in the mela- nogenic pathway responsible for the hydroxylation of L-tyrosine to 3,4-dihydroxy phenylalanine (L-DOPA) and oxidation of L-DOPA to dopaquinone.3,4 However, an excessive accumulation of the pigment could lead to serious aesthetic issues. Alterations in tyrosinase function could culminate with serious dermatological http://dx.doi.org/10.1016/j.bmcl.2015.10.071 0960-894X/Ó 2015 Elsevier Ltd. All rights reserved. ⇑ Corresponding author. Tel.: +61 426687300. E-mail address: Sini.KaranayilRadhakrishnan@student.uts.edu.au (S. Radhakrishnan). O + O R 1 H a O R 1 Scheme 1. General method for synthesis of 2,3-dihydro-1H-inden-1-one chalcones. Reagents and conditions: (a) MeOH, NaOH, 0 °C, 24 h. Table 1 Substitution pattern and tyrosinase inhibition effects of 1-indanone derivatives O R 1 R 2 R 3 R 4 Compound R1 R2 R3 R4 Yield (%) Inhibition rate (%) at 50 lM 1a H NO2 H H 95.5 22.6 ± 0.44 1b H H NO2 H 87.6 19.8 ± 0.15 1c OMe H NO2 H 64.2 25.4 ± 0.55 1d H NO2 OMe H 54.1 24.2 ± 0.33 1e OMe H OMe H 85.4 30.6 ± 0.47 1f H OMe OMe H 77.2 32.2 ± 0.11 1g H OMe H OMe 70.3 38.6 ± 0.32 2a OH H OH H 51.2 65.2 ± 0.15 2b H OH OH H 45.2 74.6 ± 0.14 2c H OH H OH 42.6 50.1 ± 0.38 Kojic acid 45.2 ± 0.22 O OMe b O OH Scheme 2. Dealkylation of methoxy chalcones. Reagents and conditions: (b) BBr3, CH2Cl2. Bioorganic & Medicinal Chemistry Letters xxx (2015) xxx–xxx Contents lists available at ScienceDirect Bioorganic & Medicinal Chemistry Letters journal homepage: www.elsevier.com/locate/bmcl Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
  • 2. disorders including melasma, ephelides (freckles), solar lentigo (age spots), and sites of actinic damage.5–7 Tyrosinase is responsi- ble for melanization in animals, cuticle formation in insects, wound healing and browning of fruits and vegetables.8–15 There- fore, tyrosinase inhibitors have potential applications in the treat- ment of dermatological disorders associated with melanin hyperpigmentation, in agriculture as bio-insecticides and also in cosmetics for whitening and depigmentation after sunburn. From a structural perspective, tyrosinase has two copper ions in its active site which play a vital role in its catalytic activity. At the active site of tyrosinase, a dioxygen molecule binds in side-on coordination between two copper ions. Each of the copper ions is coordinated by three histidines in the protein matrix. 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 produc- tion of the brown color.16,17 Several natural and synthetic tyrosinase inhibitors have been reported, including aromatic aldehydes and acids, tropolone, arbutin, flavonoids, and kojic acid.18,19 However, most of these compounds have been reported to be either toxic towards cells or have low stability towards oxygen and water.20–22 Indanones are bioactive molecules that are frequently used as precursors in the synthesis of pharmaceutical and natural product substances. Previously, indanone based curcumin analogs have been reported to have strong inhibitory effects on tyrosinase.23 Another indanone derivative, tetrahydrocurcumin, was recom- mended to be used in cosmetics as a lighting agent.24 Although indole and its derivatives are popular medicinal agents, the related heterocycles like indene and their derivatives like substituted 1-indanones (2,3-dihydro-1H-inden-1-one) have much less been exploited for their biological potential. Previously we have studied the tyrosinase inhibition of azachalcone com- pounds.25 Hence, we considered that it might be interesting to syn- thesize a series of novel 2,3-dihydro-1H-inden-1-one chalcone-like 0 20 40 60 80 100 120 0 5 10 15 20 Relativeactivity(%) [I] (µM) 2a 2b Figure 1. Inhibition effects of compounds 2a and 2b on the diphenolase activity of mushroom tyrosinase. Data are presented as means (n = 3). 0 50 100 150 200 250 0 2 4 6 8 10 12 Activity(µM/min) [E] (µg/mL) 0 1 2 3 4 0 20 40 60 80 100 120 140 160 180 0 2 4 6 8 10 12 Activity(µM/min) [E] (µg/mL) 4 1 2 3 0 2b 2a Figure 2. The inhibitory mechanism of compounds 2a and 2b on mushroom tyrosinase were reversible. The concentration of inhibitor used for curves 0–4 are 0, 0.25, 0.5, 1.0 and 2.0 lM, respectively. 2 S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
  • 3. compounds and their hydroxyl derivatives for use as depigmenta- tion agents and as anti-browning food additives. In the first step, 2,3-dihydro-1H-inden-1-one chalcone-like compounds were synthesized by the base catalyzed Claisen– Schmidt condensation of 1-indanone and an appropriate ketone in a polar solvent like methanol (Scheme 1). Some selected methoxy chalcones were then successfully dealkylated to their corresponding hydroxy compounds in the presence of boron tribromide (BBR3) (Scheme 2). In the present study, tyrosinase extracted from the edible mushroom Agaricus bisporus is used due to its easy availability and high homology with the mammalian enzyme that renders it well suited as a model for studies on melanogenesis.26 We used L-DOPA as substrate to study the effect of inhibitor compounds on the oxidation of L-DOPA by tyrosinase (diphenolase activity).27 Kojic acid, a well-known tyrosinase inhibitor served as the positive control. We have further investigated the kinetic parameters and inhibition mechanisms of active tyrosinase inhibitor compounds. Dose-dependent inhibition experiments were performed in tripli- cate to determine the IC50 of the active inhibitors 2a and 2b. In terms of the structure–activity relationships, compound 2b ((2Z)-2-(3,4-dihydroxybenzylidene)-2,3-dihydro-1H-inden-1-one) exhibited the most potent tyrosinase inhibitory activity with inhibition of 74.6%. This could be accounted to the presence of a 3,4-dihydroxy group in the B-ring that shows structural resemblance with the substrate, L-DOPA. Compound 2a with an ortho–para dihydroxy substitution on ring B showed an inhibition of 65.2% at 50 lM. The inhibition of both compounds 2a and 2b was seen to be greater than the positive control kojic acid which exhibited 45.2% inhibition at 50 lM. The inhibitory activities of these newly synthesized compounds were compared to that of kojic acid as a reference standard and the results are shown in Table 1. Results indicated that both compounds 2a and 2b could inhibit the diphenolase activity of tyrosinase in a dose-dependent manner. With increasing concentrations of inhibitors, the remaining enzyme activity decreased exponentially. The IC50 for compounds 2a and 2b was estimated to be 12.3 lM and 8.2 lM, respectively (Fig. 1). The plots of the remaining enzyme activity versus the concen- tration of enzyme at different inhibitor concentrations gave a family of straight lines, which all passed through the origin. The presence of inhibitor did not reduce the amount of enzyme, but just resulted in the inhibition of enzyme activity. The results showed that both the compounds 2a and 2b were reversible inhi- bitors of mushroom tyrosinase for oxidation of L-DOPA (Fig. 2). Compounds 1c, 2b, 2c and 2d exhibited moderate tyrosinase inhibition while compounds 1a, 1b and 1d–1f showed negligible tyrosinase inhibition in comparison with kojic acid. Previous stud- ies have recognized potent inhibitory effect of chalcones with hydroxyl groups on tyrosinase.28–34 The hydroxyl groups in com- pounds carry out the nucleophilic attack on the coppers of tyrosi- nase active site and directly involved in transferring protons during catalysis, which resulted in inactivation of tyrosinase.35,36 Further- more, results indicated that electron-donating groups contributed more to the inhibitory activity of the compounds on mushroom -200 -100 0 100 200 300 400 -4 -2 0 2 4 6 1/V(µM/min)-1 1/S (mM/L) 2a -200 -100 0 100 200 300 400 -4 -2 0 2 4 6 1/V(µM/min)-1 1/S (mM/L) 2b Figure 3. Lineweaver Burk plot for inhibition of compounds 2a and 2b on mushroom tyrosinase. Data were obtained as mean values of 1/V, the inverse of the absorbance increase at a wavelength of 492 nm per min of three independent tests with different concentrations of L-DOPA as a substrate. The concentration of compounds 2a and 2b from top to bottom is 20 lM, 5 lM, 1.25 lM and 0 lM, respectively. S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx 3 Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
  • 4. tyrosinase than the electron-withdrawing groups. Electron donat- ing groups (e.g., –OMe, –OH) on the atoms adjacent to the p system activate the aromatic ring by increasing the electron density on the ring through a resonance donating effect. In this study, we investigated in depth the tyrosinase inhibitory activities of compounds 2b and 2a. We measured the reaction rates in the presence of active inhibitors with various concentrations of L-DOPA as a substrate. As the concentrations of active inhibitors 2a and 2b increased, Km values gradually increased, but Vmax values did not change, thereby indicating the inhibitors act as competitive inhibitors of mushroom tyrosinase (Fig. 3). The inhibition kinetics were illustrated by Dixon plots, which were obtained by plotting 1/V versus [I] with varying concentra- tions of substrate. Dixon plots gave a family of straight lines pass- ing through the same point at the second quadrant, giving the inhibition constant (Ki) (Fig. 4). The Ki value for the compounds 2a and 2b estimated from this Dixon plot was 10.3 lM and 8.7 lM, respectively. A comparison of the Ki values of the compounds with that of kojic acid revealed that they possess much higher affinity to tyrosinase than kojic acid (Table 2). To summarize, in this study, we synthesized novel 2,3-dihydro- 1H-inden-1-one (1-indanone) chalcone-like compounds and their hydroxy derivatives, and studied their inhibition on the diphenolase activity of mushroom tyrosinase. Hydroxy substituted 1-indanone chalcone-like compounds 2a and 2b were found to be significantly more potent than kojic acid with their IC50 values of 12.3 lM and 8.2 lM, respectively. Both the compounds exhibited reversible competitive inhibition. Compound 2b with a 3,4-dihy- droxy group in the B-ring had structural resemblance to the substrate L-DOPA. This leads to the competitive displacement of L-DOPA from the active site of the enzyme tyrosinase in a lock- and key model. In addition, the presence of the hydroxy group at the 4-position (ring B) is indispensable for significant tyrosinase inhibition in this class of compounds. These preliminary findings justify pursuing further work on hydroxy substituted 1-indanone chalcone-like compounds to serve as a potential scaffold for the future development of active tyrosinase inhibitors. Notes: 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 -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 -30 -20 -10 0 10 20 30 40 -40 -30 -20 -10 0 10 20 30 1/V(µM/min)-1 [inhibitor] µM 2a Figure 4. Dixon plot for the inhibitory effect of compounds 2a and 2b on L-DOPA oxidation catalyzed by mushroom tyrosinase. The inhibitor concentrations were 0, 10 lM and 20 lM, respectively. The L-DOPA concentrations were 200, 400 and 600 lM. Table 2 Effect on mushroom polyphenol oxidase activity and kinetic analysis of compounds Compound Type of inhibition IC50 (lM) RA* Ki # (lM) 2a Competitive 12.3 ± 0.22 2.23F 10.3 2b Competitive 8.2 ± 0.44 3.35F 8.7 Kojic acid Competitive 27.5 ± 0.56 12.2 # Values were measured at 5 lM of active compounds and Ki is the (inhibitor constant). * RA is the relative inhibitory activity compared to the standard kojic acid where 1.0F means one time activity of kojic acid. 4 S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071
  • 5. chemical shifts (o) are quoted in parts per million downfield from TMS and coupling constants (J) are given in Hz. Abbreviations used in the splitting pattern were as follows: s = singlet, d = doublet, t = triplet, m = multiplet. 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 purchased from Sigma–Aldrich Chemical Co. General method for the synthesis of 2,3-dihydro-1H-inden-1-one (1-indanone) chalcone-like compounds (1a–1g): To a stirred solution of 1-indanone (2.0 mmol) and the corresponding aldehyde (2.0 mmol) in methanol (6 mL) at room temperature was added freshly pulverized sodium hydroxide (160 mg, 4.0 mmol). The solution was stirred overnight (monitored by TLC) and then poured onto crushed ice. Once all the ice had melted, pH was adjusted to 5 using hydrochloric acid (5 M). The resulting solution was then extracted with ethyl acetate, washed with brine and filtered after drying with anhydrous sodium sulfate. The solution was then concentrated with a rotary evaporator under vacuum to give the respective compound. Method for the synthesis of hydroxy indanone compounds (2a–2c): A solution of BBr3 (2.5 mL for each methoxy group) was added to a cooled solution of the corresponding methoxy chalcone (1 mmol) in CH2Cl2 under argon. The cooling bath was removed and the dark solution was warmed to rt and stirred for 1–5 h. The dark solution was then poured into ice water and filtered. The aqueous layer was extracted with chloroform twice. The organic extract was washed with water followed by brine and anhydrous sodium sulfate. This was then rotary evaporated to give the respective product. Enzyme activity assay: The mushroom tyrosinase inhibition activity of 1-indanone chalcone-like compounds was measured using L-DOPA as substrate. The activity assay used 3 ml of reaction medium containing 0.5 mM L-DOPA in 50 mM Na2HPO4–NaH2PO4 buffer (pH 6.8). The final concentrations of tyrosinase was 3.33 mg/ml. The substrate reaction progress curve was analyzed to obtain the reaction rate constants. The reaction was carried out at 30 °C and pH 6.8. Determination of the inhibition type of compounds 2a and 2b 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 lL; 0.2 mg mLÀ1 ) was incubated with 50 lL of various concen- trations of enzyme substrate and 50 lL of phosphate buffer, and then 50 lL of different concentrations of tested samples 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 ana- lyzed by Dixon plot, which is a graphical method plot of 1/enzyme velocity (1/V) versus inhibitor concentration (I) with varying con- centrations of the substrate. Supplementary data Supplementary data associated with this article can be found, in the online version, at http://dx.doi.org/10.1016/j.bmcl.2015.10. 071. References and notes 1. Hearing, V. J. Methods Enzymol. 1987, 142, 154. 2. Spritz, R. A.; Hearing, V. J. Adv. Hum. Genet. 1994, 22, 1. 3. Cooksey, C. J.; Garratt, P. J.; Land, E. J.; Pavel, S.; Ramsden, C. A.; Riley, P. A.; Smit, N. P. J. Biol. Chem. 1997, 272, 26226. 4. Kubo, I.; Kinst-Hori, I.; Chaudhuri, S. K.; Kubo, Y.; Ogura, Y. T. Bioorg. Med. Chem. 2000, 8, 1749. 5. Iozumi, K.; Hoganson, G. E.; Pennella, R.; Everett, M. A.; Fuller, B. B. J. Invest. Dermatol. 1993, 100, 806. 6. Ando, H.; Kondoh, H.; Ichihashi, M.; Hearing, V. J. Invest. Dermatol. 2007, 127, 751. 7. Wang, H. M.; Chen, C. Y.; Wen, Z. H. Exp. Dermatol. 2011, 20, 242. 8. Olivares, C.; Solano, F. Pigment Cell Melanoma Res. 2009, 22, 750. 9. Jimbow, K.; Park, J. S.; Kato, F.; Hirosaki, K.; Toyofuku, K.; Hua, C.; Yamashita, T. Pigment Cell Res. 2000, 13, 222. 10. Kim, Y. J.; Uyama, H. Cell. Mol. Life Sci. 2005, 62, 1707. 11. Rescigno, A.; Sollai, F.; Pisu, B.; Rinaldi, A.; Sanjust, E. J. Enzyme Inhib. Med. Chem. 2002, 17, 207. 12. Kanost, M. R.; Jiang, H.; Yu, X. Q. Immunol. Rev. 2004, 198, 97. 13. Lai, S. C.; Chen, C. C.; Hou, R. R. J. Med. Entomol. 2002, 39, 266. 14. Guerrero, A.; Rosell, G. Curr. Med. Chem. 2005, 12, 461. 15. Gorman, M. J.; An, C.; Kanost, M. R. Insect Biochem. Mol. Biol. 2007, 37, 1327. 16. Sanchez-Ferrer, A.; Rodriguez-Lopez, J. N.; Garcia-Canavas, F.; Garcia-Carmona, F. Biochim. Biophys. Acta 1995, 1, 1247. 17. Decker, H.; Schweikardt, T.; Nillius, D.; Salzbrunn, U.; Jaenicke, E.; Tuczek, F. Gene 2007, 398, 183. 18. Chen, Q. X.; Song, K. K.; Qiu, L.; Liu, X. D.; Huang, H.; Guo, H. Y. Food Chem. 2005, 91, 269. 19. Xie, L. P.; Chen, Q. X.; Huang, H.; Wang, H. Z.; Zhang, R. Q. Biochemistry 2003, 68, 487. 20. Burnett, C. L.; Bergfeld, W. F.; Belsito, D. V.; Hill, R. A.; Klaassen, C. D.; Liebler, D. C.; Marks, J. G.; Shank, R. C.; Slaga, T. J.; Snyder, P. W.; Andersen, F. A. Int. J. Toxicol. 2010, 29, 244. 21. Draelos, Z. D. Dermatol. Ther. 2007, 20, 308. 22. Loizzo, M. R.; Tundiz, T.; Menichini, F. Compr. Rev. Food. Sci. F 2012, 11, 378. 23. Sugimoto, H. Pure Appl. Chem. 1999, 71, 2031. 24. Asawanonda, P.; Klahan, S. O. Photomed. Laser Surg. 2010, 28, 679. 25. Radhakrishnan, K. S.; Shimmon, R.; Conn, C.; Baker, T. A. Bioorg. Med. Chem. Lett. 2015, 25, 1753. 26. Song, K. K.; Chen, Q. X.; Wang, Q.; Qiu, L.; Huang, H. J. Enzyme Inhib. Med. Chem. 2005, 20, 239. 27. Gerdemann, C.; Eicken, C.; Krebs, B. Acc. Chem. Res. 2002, 35, 183. 28. Yan, Q.; Cao, R.; Yi, W.; Yu, L.; Chen, Z.; Ma, L.; Song, H. Bioorg. Med. Chem. Lett. 2009, 19, 4055. 29. Yamazaki, Y.; Kawano, Y.; Yamanaka, A.; Maruyama, S. Bioorg. Med. Chem. Lett. 2009, 19, 4178. 30. Kanade, S. R.; Suhas, V. L.; Chandra, N.; Gowda, L. R. FEBS J. 2007, 274, 4177. 31. Kim, D.; Park, J.; Kim, J.; Han, C.; Yoon, J.; Kim, N.; Seo, J.; Lee, C. J. Agric. Food Chem. 2006, 54, 935. 32. Shiino, M.; Watanabe, Y.; Umezawa, K. Bioorg. Chem. 2003, 31, 129. 33. Shiino, M.; Watanabe, Y.; Umezawa, K. Bioorg. Med. Chem. 2001, 9, 1233. 34. Yokota, T.; Nishio, H.; Kubota, Y.; Mizoguchi, M. Pigment Cell Res. 1998, 11, 355. 35. Mu~noz-Mu~noz, J. L.; Garcia-Molina, F.; Garcia-Ruiz, P. A.; Molina-Alarcon, M.; Tudela, J.; Garcia-Canovas, F.; Rodriguez-Lopez, J. N. Biochem. J. 2008, 416, 431. 36. Espin, J. C.; Varon, R.; Fenoll, L. G.; Gilabert, M. A.; Garcia-Ruiz, P. A.; Tudela, J.; Garcia-Canovas, F. Eur. J. Biochem. 2000, 267, 1270. S. Radhakrishnan et al. / Bioorg. Med. Chem. Lett. xxx (2015) xxx–xxx 5 Please cite this article in press as: Radhakrishnan, S.; et al. Bioorg. Med. Chem. Lett. (2015), http://dx.doi.org/10.1016/j.bmcl.2015.10.071