7_Hydroxycoumarin_modulates_the_oxidativ

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Chemico-Biological Interactions 206 (2013) 63–75

Contents lists available at ScienceDirect

Chemico-Biological Interactions
journal homepage: www.elsevier.com/locate/chembioint

7-Hydroxycoumarin modulates the oxidative metabolism, degranulation


and microbial killing of human neutrophils
Luciana M. Kabeya a,⇑, Carolina N. Fuzissaki a, Silvia H. Taleb-Contini a,1, Ana Maria da C. Ferreira b,
Zeki Naal a, Everton O.L. Santos a, Andréa S.G. Figueiredo-Rinhel a, Ana Elisa C.S. Azzolini a,
Roberta B. Vermelho a, Alberto Malvezzi b,2, Antonia T. -do Amaral b, João Luis C. Lopes a,
Yara Maria Lucisano-Valim a
a
Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto da Universidade de São Paulo. Av. do Café s/n, 14040-903 Ribeirão Preto, SP, Brazil
b
Departamento de Química Fundamental, Instituto de Química da Universidade de São Paulo. Av. Prof. Lineu Prestes 748, 05508-900 São Paulo, SP, Brazil

a r t i c l e i n f o a b s t r a c t

Article history: In the present study, we assessed whether 7-hydroxycoumarin (umbelliferone), 7-hydroxy-4-methyl-
Received 16 April 2013 coumarin, and their acetylated analogs modulate some of the effector functions of human neutrophils
Received in revised form 14 July 2013 and display antioxidant activity. These compounds decreased the ability of neutrophils to generate super-
Accepted 17 August 2013
oxide anion, release primary granule enzymes, and kill Candida albicans. Cytotoxicity did not mediate
Available online 28 August 2013
their inhibitory effect, at least under the assessed conditions. These coumarins scavenged hypochlorous
acid and protected ascorbic acid from electrochemical oxidation in cell-free systems. On the other hand,
Keywords:
the four coumarins increased the luminol-enhanced chemiluminescence of human neutrophils stimu-
7-Hydroxycoumarin
Neutrophil
lated with phorbol-12-myristate-13-acetate and serum-opsonized zymosan. Oxidation of the hydroxyl-
Degranulation ated coumarins by the neutrophil myeloperoxidase produced highly reactive coumarin radical
Myeloperoxidase intermediates, which mediated the prooxidant effect observed in the luminol-enhanced chemilumines-
Candida albicans cence assay. These species also oxidized ascorbic acid and the spin traps a-(4-pyridyl-1-oxide)-N-tert-
Prooxidant butylnitrone and 5-dimethyl-1-pyrroline-N-oxide. Therefore, 7-hydroxycoumarin and the derivatives
investigated here were able to modulate the effector functions of human neutrophils and scavenge reac-
tive oxidizing species; they also generated reactive coumarin derivatives in the presence of myeloperox-
idase. Acetylation of the free hydroxyl group, but not addition of the 4-methyl group, suppressed the
biological effects of 7-hydroxycoumarin. These findings help clarify how 7-hydroxycoumarin acts on neu-
trophils to produce relevant anti-inflammatory effects.
Ó 2013 Elsevier Ireland Ltd. All rights reserved.

1. Introduction cells. These molecules help regulate the initiation of specific T


and B cell immunity and the resolution phase of inflammation
Neutrophils are professional killers of invading microorganisms [1–3]. Thus, modulating the effector functions of neutrophils is a
and also contribute to the regulation of inflammatory processes. promising therapeutic strategy to manage inflammation.
These cells secrete types of chemokines, cytokines, lipid mediators, Coumarin and its derivatives have arisen as promising natural
granule proteins, and reactive oxygen species (ROS) that serve as products with potent anti-inflammatory effect and low toxicity
signaling molecules for subsequent recruitment of inflammatory to humans [4–8]. They constitute a large class of phenolic

Abbreviations: AAPV, methoxy-succinyl-Ala-Ala-Pro-Val-chloromethylketone; ABAH, 4-aminobenzoic acid hydrazide; Asc, ascorbic acid; CL, chemiluminescence; CL-luc,
lucigenin-enhanced chemiluminescence; CL-lum, luminol-enhanced chemiluminescence; DMPO, 5-dimethyl-1-pyrroline-N-oxide; DMPOX, 5,5-dimethyl-1-pyrrolidone-2-
oxyl-1; DMSO, dimethyl sulfoxide; DPPH, 1,1-diphenyl-2-picrylhydrazyl radical; EPR, electron paramagnetic resonance; n-fMLP, n-formyl-methionyl-leucyl-phenylalanine;
HBSS, Hank’s balanced saline solution; HBSS-gel, Hank’s balanced saline solution supplemented with gelatin; HRP, horseradish peroxidase; IC50, concentration inhibiting a
biological response by 50%; LDH, lactate dehydrogenase; MPO, myeloperoxidase; MTT, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide; NADPH, reduced
form of nicotinamide adenine dinucleotide phosphate; PI, propidium iodide; PKC, protein kinase C; PMA, phorbol-12-myristate-13-acetate; 4-POBN, a-(4-pyridyl-1-oxide)-N-
tert-butylnitrone; ROS, reactive oxygen species; SAAVNA, N-succinyl-Ala-Ala-Val-p-nitroanilide; SOD, superoxide dismutase; SOZ, serum-opsonized zymosan.
⇑ Corresponding author. Address: Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo. Av. do Café s/n,
Bairro Monte Alegre, 14040-903 Ribeirão Preto, SP, Brazil. Tel.: +55 16 36024212; fax: +55 16 36024880.
E-mail address: luciana_kabeya@yahoo.com.br (L.M. Kabeya).
1
Present address: Unidade de Biotecnologia, Universidade de Ribeirão Preto (UNAERP), Avenida Costábile Romano 2201, Bairro Ribeirânea, 14096-380 Ribeirão Preto, SP, Brazil.
2
Present address: Universidade Nove de Julho (UNINOVE), Avenida Dr. Adolpho Pinto 109, Barra Funda, São Paulo, SP 01156-050, Brazil.

0009-2797/$ - see front matter Ó 2013 Elsevier Ireland Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.cbi.2013.08.010
64 L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75

compounds produced by plants of different botanical families, VI-A, EC.1.11.1.7), luminol (5-amine-2,3-dihydro-1,4-phtalazinedi-
primarily in Angiospermae [9]. The coumarin (1,2-benzopyrone; one), lucigenin (N,N’-dimethyl-9,90 -biacridinium dinitrate), 3-(4,5-
5,6-benzo-a-pyrone) has been clinically used to treat cancer, dimethyl-2-thiazolyl)-2,5- diphenyl-2H-tetrazolium bromide
lymphoedema, venous insufficiency and chronic infections [5–8]. (MTT), phorbol-12-myristate-13-acetate (PMA), superoxide dis-
Coumarin alone is ineffective in treating some diseases, but it mutase (SOD), trizma hydrochloride (Tris(hydroxymethyl)amino-
can increase the beneficial effects of other compounds in combina- methane hydrochloride), 3,30 ,5,50 -tetramethylbenzidine, Triton
tion therapy [8]. However, it has a short half-life in humans due to X-100, Trypan blue, and Zymosan A were purchased from Sigma–
hepatic metabolism (70–90%) to 7-hydroxycoumarin (Fig. 1) and Aldrich (St. Louis, MO, USA). Dimethyl sulfoxide (DMSO), ethanol,
its glucuronide. Coumarin is considered a prodrug and 7-hydroxy- n-octanol, hydrogen peroxide, n-formyl-methionyl-leucyl-phenyl-
coumarin is the bioactive molecule [6,8]. alanine (n-fMLP), N-succinyl-Ala-Ala-Val-p-nitroanilide (SAAVNA),
7-Hydroxycoumarin, also known as umbelliferone, occurs in methoxy-succinyl-Ala-Ala-Pro-Val-chloromethylketone (AAPV),
plants and edible fruits and roots, such as golden apple, bitter or- human leukocyte myeloperoxidase (MPO, EC 1.7.1.11) and 4-ami-
ange, carrot, coriander, garden angelica, and mouse-ear hawkweed nobenzoic acid hydrazide (ABAH) were obtained from Merck
[10–11]. This coumarin displays a wide range of beneficial bioac- (Darmstadt, Germany). We acquired the following products from
tivities: it reduces lymphoedema in humans [6] and exerts anti- different suppliers: gelatin (microbiological grade, Difco Laborato-
hyperglycemic [12], bronchodilating [10], antinociceptive [13], ries, Detroit, MN, USA), propylene glycol (Synth, Diadema, SP,
and antiedematogenic [4,14,15] effects in rats. The in vitro and Brazil), RPMI 1640 medium (Cultilab, Campinas, SP, Brazil),
in vivo antioxidant, anti-inflammatory and immunomodulatory ef- Sabouraud’s dextrose agar (Acumedia, Michigan, USA), LDH Liqui-
fects of 7-hydroxycoumarin have been recently reported form™ kit (Labtest Diagnostica, Lagoa Santa, MG, Brazil) and
[4,5,11,16–22]. This compound inhibits migration of neutrophils APOPTEST™-FITC (propidium iodide (PI) + Annexin V stained with
and eosinophils [10], degranulation of mast cells [15], release of fluorescein isothiocyanate) apoptosis detection kit (Dako, Glostrup,
NO by macrophages [19–21], release of cytokines [10,13], biosyn- Denmark). All the other chemicals and solvents used in this work
thesis of prostaglandin, and activity of cycloxygenase-2 and were of analytical grade and purchased from commercial sources.
5-lipoxygenase [4,5,14]. In addition, 7-hydroxycoumarin activates
the protective immune responses of mice against influenza virus 2.2. Coumarins
[19] and Salmonella typhimurium[22].
However, there are few reports on the action of 7-hydroxy- 7-Hydroxycoumarin (1) was purchased from Sigma–Aldrich
coumarin on neutrophils. 7-Hydroxycoumarin and 7-hydroxy (St Louis, MO, USA). It was used as raw material to synthesize 7-hy-
-4-methylcoumarin inhibit the generation of O 2 by zymosan- droxy-4-methylcoumarin (2), 7-acetoxycoumarin (3), and 7-acet-
stimulated rabbit neutrophils [23], phorbol-12-myristate-13 oxy-4-methylcoumarin (4), by the methods described by Vogel
-acetate (PMA)-stimulated human neutrophils, and n-formyl- [24] and Lopes et al. [25], with slight modifications. The purity of
methionyl-leucyl-phenylalanine (fMLP)-stimulated rat leukocytes all the compounds was 97–100%. Their chemical structures are
[5]. 7-Hydroxycoumarin also inhibits the migration of neutrophils illustrated in Fig. 1.
in mouse models of inflammation [10,13]. Therefore, the high po-
tential of 7-hydroxycoumarin for the development of an effective 2.3. Isolation of human neutrophils
anti-inflammatory drug and the fundamental contribution of
neutrophils to the regulation of inflammatory processes have moti- Twenty adult volunteers who met the criteria described by Pau-
vated us to evaluate the modulator effect of 7-hydroxycoumarin, la et al. [26] were recruited for this study. The experimental proce-
7-hydroxy-4-methylcoumarin, and their acetylated analogs dures were approved by the local Research Ethics Committee on
(Fig. 1) on some effector functions of human neutrophils. In partic- Human Experimentation (CEP/FCFRP-USP, protocol 96), according
ular, we investigated their effects on the oxidative metabolism, to the rules of the 196/96 Resolution of the Brazilian National
degranulation, microbial killing ability, and myeloperoxidase and Health Council and the Helsinki Declaration of 1975 (revised in
elastase activity of neutrophils. We also evaluated their physico- 2008).
chemical properties, antioxidant activity, and HOCl scavenging po- Blood was collected by venous puncture and diluted 1:2 in
tential in cell-free systems. Alsever solution (anticoagulant). Neutrophils were isolated using
sterile and lipopolysaccharide (LPS)-free solutions, as previously
described [26]. The cell pellets were suspended in Hanks balanced
2. Materials and methods saline solution (HBSS) containing 1 mg mL-1 gelatin (HBSS-gel).
The cell preparations contained 90–95% neutrophils with viability
2.1. Chemicals higher than 95%, as established by exclusion with Trypan Blue.

L-Ascorbic acid (Asc), cytochalasin B, 1,1-diphenyl-2 2.4. Generation of ROS by neutrophils


-picrylhydrazyl radical (DPPH), N,N-dimethylformamide, 5-di-
methyl-1-pyrroline-N-oxide (DMPO), a-(4-pyridyl-1-oxide) The generation of ROS by neutrophils was measured as de-
-N-tert-butylnitrone (4-POBN), horseradish peroxidase (HRP, type scribed previously [26]. Zymosan particles were prepared and
opsonized with normal human serum as described by Kanashiro
et al. [27]. Serum-opsonized zymosan (SOZ) was suspended in
HBSS for use (10 mg mL1). A 10 mM PMA stock solution in
DMSO was diluted 1:1000 in HBSS prior to use [26].
Briefly, the reaction mixtures (0.5 mL) containing neutrophils
(1  106 cells mL1), the chemiluminescent probe (100 lM luminol
or 100 lM lucigenin) and a coumarin (0.01–100 lM), ABAH
(1–200 lM) or DMSO (0.1% v/v, control) were incubated for
3 min, at 37 °C. The reaction was initiated by adding SOZ
(1 mg mL1) or PMA (0.1 lM) and the luminol- or lucigenin-
Fig. 1. Chemical structure of coumarins. enhanced chemiluminescence (CL-lum and CL-luc, respectively)
L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75 65

was measured for 15 min, at 37 °C, in a luminometer (Auto Lumat 2.8. Evaluation of the cytotoxicity
LB953, EG&G Berthold, Bad Wildbad, Germany). The light emission
was recorded in c.p.m. (photons counts per minute). Background The cytotoxic effect of coumarins on neutrophils was evaluated
CL produced by non-stimulated cells was subtracted from all the as previously described [30]. Briefly, aliquots of neutrophils
samples. The integrated areas of CL were determined and used to (1  106 cells mL1) were incubated with one of the coumarins
calculate the percentage of CL inhibition or increase promoted by (200 lM), DMSO (0.1% v/v, vehicle), HBSS (non-treated cells), or
the tested compound. Triton X-100 (0.2% v/v, positive control) for 20 min or 60 min, at
37 °C. The cell pellets were immediately suspended in HBSS-gel
after centrifugation (755g, 10 min, 4 °C), and cellular viability
2.5. Degranulation assay was determined using the Trypan Blue exclusion test, by counting
a total of 200 cells for each sample. The activity of lactate dehydro-
Degranulation of neutrophils was evaluated as described previ- genase (LDH) released into the supernatant was measured with the
ously [28], using elastase as marker. Briefly, aliquots of neutrophils LDH Liquiform™ test kit.
(1  106 cells mL1) were incubated in 96-well microplates with a Neutrophils (1  106 cells mL1) were treated with coumarin 1
coumarin (100 lM), DMSO (0.1% v/v, vehicle), or HBSS (control) for (100 lM) for 60 min, at 37 °C. The cell pellets were immediately
20 min or 60 min, at 37 °C. Then, cytochalasin B (1 lM) was added suspended in Annexin V-FITC binding buffer after centrifugation
to the mixture and incubated for 10 min, at 37 °C, before adding n- (440g, 10 min, 4 °C) and kept on ice for 10 min in the dark. PI
fMLP (1 lM) to trigger degranulation. After 30 min of incubation at was added to the samples, and they were immediately analyzed
37 °C, the elastase substrate SAAVNA (330 lM) was added to the in the BD FACSCanto flow cytometer (Becton, Dickinson and Com-
medium and the mixture was incubated for further 30 min, at pany; Franklin Lakes, NJ, USA). Data from 10,000 events were col-
37 °C. The absorbance was recorded at 405 nm in a microplate lected and analyzed by the BD FACSDiva Software. We followed the
reader (EIA Multi-well Reader II, Sigma Diagnostics, St. Louis, instructions of the manufacturer of the APOPTEST™-FITC apoptosis
MO, USA). The difference between the absorbance readings before detection kit to perform this assay.
and after addition of SAAVNA was used to calculate the percentage
of degranulation elicited by each sample. 2.9. HOCl scavenging assay

The HOCl scavenging potential of coumarins was assessed as


2.6. Elastase activity of human neutrophils described by Andrade et al. [31]. The working solution of one of
the coumarins (0.05–50 lM) or propylene glycol (0.1% v/v, control)
The elastase-rich supernatant was obtained from human neu- was added to HOCl (50 lM) and allowed to react for 10 min. The
trophils treated with cytochalasin B and n-fMLP, as described by residual HOCl was detected by rapidly mixing the developing re-
Kanashiro et al. [28]. Aliquots of the supernatant were mixed with agent (2 mM 3,30 ,5,50 -tetramethylbenzidine in 400 mM acetate
one of the coumarins (100 lM), DMSO (0.1% v/v, vehicle) or HBSS buffer pH 5.4 containing 10% N,N-dimethylformamide and
(control), and incubated for 20 min or 60 min, at 37 °C. Then SAAV- 100 lM KI). The absorbance was recorded at 650 nm in a plate
NA (330 lM) was added to all the samples, and the absorbance of reader after 5 min of incubation. All the reaction steps were con-
the product p-nitroaniline was recorded at 405 nm in a plate read- ducted in the dark at 25 °C.
er, after 30 min of incubation at 37 °C. Appropriate blanks without
the substrate were subtracted from all the samples. 2.10. DPPH scavenging assay

The DPPH free radical scavenging potential of coumarins was


2.7. Microbial killing by neutrophils examined by the method of Blois [32], with modifications. The
DPPH stock solution was prepared in ethanol and diluted 6:4 in
This assay was performed in 96-well microplates, according to 40 mM acetate buffer pH 5.5 for use. The DPPH working
the method described by Mosmann [29], with modifications. Star- solution (100 lM) was incubated with one of the coumarins
ter cultures of Candida albicans (ATCC 64548) were grown for 48 h, (0.1–100 lM), DMSO (0.1% v/v, vehicle), or HBSS (control) for
at 37 °C, in Sabouraud’s agar containing 1% chloramphenicol. The 5 min, at 25 °C, and the absorbance was recorded at 517 nm. The
cells were harvested, washed with sterile 0.15 M NaCl (3000g, difference between absorbance readings before and after sample
5 min, 25 °C), and suspended in RPMI medium. The suspension addition was used to calculate the percentage of DPPH reduction
containing C. albicans was kept on ice-bath until use. by each sample.
Aliquots of neutrophils (2  106 cells mL1) were treated with
one of the coumarins (1–100 lM), ABAH (50 lM), DMSO (0.02%) 2.11. MPO-H2O2-luminol chemiluminescence assay
or HBSS (control), for 30 min, at 37 °C, before adding the suspen-
sion of C. albicans (1  106 CFU mL1) to the reaction mixture. After The enzymatic activity in the MPO stock solution was deter-
2-h incubation at 37 °C in a shaker, the plates were centrifuged, mined as described by Marquez et al. [33]. The assay buffers were:
and the pellets were treated with 1% (v/v) Triton X-100 for PBS pH 7.4 (10 mM sodium phosphate, 140 mM NaCl, 10 mM KCl)
10 min, at 25 °C. The plates were centrifuged again, and the pellets and 50 mM phosphate buffer pH 7.4 (chloride-free medium). The
were washed three times with 0.15 M NaCl. MTT (0.25 mg mL1 in effect of the coumarins on the MPO-H2O2-luminol reaction was
RPMI medium) was added to all the wells, and the plates were evaluated as described by Kitagawa et al. [34], with modifications.
incubated in a shaker for 4 h, at 37 °C, in the dark. Next, the plates Reaction mixtures (0.5 mL) containing the assay buffer, luminol
were centrifuged, and the formazan crystals were dissolved in (20 lM), MPO (50 mU.mL1), and one of the coumarins
DMSO. The absorbance was recorded in a plate reader at 545 nm (0.01–10 lM) or DMSO (0.01% v/v, control), were incubated at
after 10-min incubation at 25 °C. All the centrifugation steps de- 37 °C for 2 min. The reaction was initiated with H2O2 (50 lM),
scribed in this paragraph were carried out at 3220g, for 10 min, and the CL was measured in a luminometer for 30 min, at 37 °C.
at 25 °C. Controls in the absence of neutrophils represent 100% of The integrated chemiluminescence area was used to calculate the
C. albicans viability (0% of killing). percentage of CL increase promoted by the tested compounds.
66 L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75

2.12. Oxidation of coumarins by MPO-H2O2 25 ± 1 °C. The concentration of the tested compounds in the
aqueous phase was measured spectrophotometrically at 325 nm
The coumarin solution (50 lM) or DMSO (0.1% v/v, control) was (coumarins 1 and 2: e = 11,475 and 11,055 M1 cm1, respectively)
mixed with MPO (0.5 U mL1). A small aliquot of H2O2 (50 lM) was or 275 nm (coumarins 3 and 4: e = 10,039 and 9335 M1 cm1,
added to the reaction mixture, and the spectra (200–600 nm) were respectively).
recorded in quartz cuvettes (1-cm light path) at different times
(0.5–20 min) of the reaction at 25 °C (U3501 spectrophotometer; 2.17. Data analysis
Hitachi Instruments, Tokyo, Japan). The assay buffers were PBS
pH 7.4 and 50 mM phosphate buffer pH 7.4. Experimental data were processed and analyzed using the
GraphPad Prism Software (version 3.00 for Windows, GraphPad
2.13. Oxidation of ascorbic acid by HRP-H2O2 Software Inc., San Diego, CA, USA). Statistical analysis was per-
formed by Analysis of Variance (ANOVA) followed by the Tukey’s
Aliquots of HRP solution (20 U mL1) were mixed with ascorbic test. A value of p < 0.05 was considered significant.
acid (Asc, 0.4 mM) and one of the coumarins (0–120 lM) or DMSO
(0.8% v/v; control). The reaction mixture was immediately trans-
ferred to a 200-lL flat quartz cell after addition of H2O2 3. Results
(0.4 mM). The EPR spectra were registered using modulation
amplitude of 1 G and one scan, at 22 °C. 3.1. Modulation of the ROS generation by neutrophils

2.14. Electron paramagnetic resonance (EPR) spin trap measurements We evaluated the modulator effect of the four coumarins (1–4)
on the production of O2 and total ROS by SOZ- or PMA-stimulated

DMPO was purified by recommended methods; it was treated human neutrophils using the lucigenin-(CL-luc) and luminol-
with charcoal [35] or freshly distilled [36] and stored at 80 °C. (CL-lum) enhanced chemiluminescence assays, respectively.
The coumarin stock solutions were prepared in DMSO; all the other All the tested coumarins inhibited the PMA- and SOZ-stimu-
reactants were prepared in 0.1 M sodium phosphate buffer pH 7.4. lated neutrophil CL-luc in a concentration-dependent manner
Aliquots of HRP solution (20 U mL1) were mixed with one of (Fig. 2A–D). They inhibited the SOZ-stimulated CL-luc twice as
the coumarins (0–120 lM) or DMSO (0.8% v/v), and one of the spin effectively as they inhibited the PMA-stimulated CL-luc. The
traps: DMPO (80 mM) or 4-POBN (100 mM). The reaction was hydroxylated coumarins 1 and 2 were more effective to inhibit
started by adding H2O2 (0.5 mM); the reaction mixture was imme- both the PMA- and SOZ-stimulated CL-luc than their acetylated
diately transferred to a 200-lL flat quartz cell. counterparts 3 and 4.
The EPR signals were registered at different time points of the In contrast, the four coumarins increased the PMA- and SOZ-
reaction course (0.5–30 min), at 22 °C, using the Bruker EMX spec- stimulated CL-lum of neutrophils (Fig. 2F–K). The hydroxylated
trometer operating at the X-band (9.5 GHz), with modulation fre- coumarins 1 and 2 had similar enhancing effects, which were sig-
quency of 100 kHz, microwave power of 20 mW, and modulation nificantly higher than the effect of their acetylated analogs (Fig. 2F
amplitude of 1 G. Ten scans were accumulated for each sample, and I). The percentage enhancement of the PMA-stimulated
to obtain the final spectra. The Win-EPR software package (Bruker, CL-lum of neutrophils and the peak height of the CL-lum response
USA) was used to analyze the spectra by double integration and increased as a function of the concentration of the coumarin (Fig. 2I
simulate their hyperfine coupling constants. and K). On the other hand, coumarins 1 and 2 had a biphasic effect
on the SOZ-stimulated CL-lum: the CL-lum response increased at
2.15. Measurement of redox potential by cyclic voltammetry concentrations up 10 lM but decreased at higher concentrations
(Fig. 2F). The kinetics of CL-lum produced by the SOZ-stimulated
Aliquots of the sample – coumarin (0.1–0.5 mM), luminol neutrophils changed significantly: the peak height decreased, and
(0.5 mM), ascorbic acid (Asc, 0.1–0.5 mM) or DMSO (0.1% v/v) – the time necessary to reach the maximum CL-lum response was
were added to PBS pH 7.4 previously bubbled with oxygen-free longer in the presence of increasing concentrations of the couma-
argon for 10 min. Cyclic voltammograms were recorded by a rins (Fig. 2H).
potentiostat (model MQI 12/8PCC, Microquímica, Brazil) at a ABAH, a specific MPO inhibitor, suppressed the CL-lum of neu-
sweep rate of 100 mV s1, at 25 °C. Conventional electrochemical trophils in a concentration-dependent way; the maximum inhibi-
cells were employed; a disk glassy carbon electrode (diame- tory effect lay around 60% at 50 lM. ABAH did not inhibit the
ter = 2 mm), a platinum wire, and sodium chloride saturated sil- CL-luc of neutrophils, indicating its specificity for MPO (Fig. 2E).
ver/silver chloride [Ag/AgCl/NaCl(sat)] were used as the working, This compound mitigated the effect of coumarins on the
counter, and reference electrodes, respectively. The argon gas flow PMA- and SOZ-stimulated CL-lum of neutrophils (Fig. 2G and J).
was kept near the solution surface when registering the cyclic Therefore, the fact that coumarins enhanced CL-lum depended on
voltammogram. the activity of MPO.

2.16. Physicochemical properties of coumarins 3.2. Oxidation of coumarins by MPO

The molecular volume of the tested compounds was calcu- We used cell-free systems to evaluate the oxidation of couma-
lated using the SYBYL Molecular Modeling System (version 6.7, rins by MPO-H2O2. The hydroxylated coumarins (1, 2) increased
2001, Tripos Associates Inc., St. Louis, MO, USA). The theoretical the oxidation of luminol in a concentration-dependent manner,
partition coefficient value (ClogP) was determined by the ClogP but their acetylated counterparts (3, 4) did not interfere signifi-
for Windows (version 1.0.0, 1995, ByoByte Corp., USA). The cantly in this reaction (Fig. 3A). In the chloride-free reaction med-
apparent partition coefficient (LogPapp) was obtained by the ium, the prooxidant effect of coumarins 1 and 2 was fourfold
shake-flask method, according to the procedure described by higher than the effect observed in the presence of chloride
Amaral et al. [37]. The partition system consisted of saturated (Fig. 3B). However, in the absence of luminol, the hydroxylated
n-octanol and trizma buffer pH 7.4 with 0.1 M ionic strength, ad- compounds were oxidized a little faster in the reaction medium
justed with NaCl. Room temperature was controlled and set to containing chloride (Fig. 3D).
L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75 67

Fig. 2. Modulation of the SOZ- and PMA-stimulated oxidative metabolism of human neutrophils by 7-hydroxycoumarin derivatives. (A–D): Concentration-dependent
2 by neutrophils stimulated with SOZ (A, B) and PMA (C, D), assessed by the lucigenin-amplified chemiluminescence (CL-luc) assay [(A) and
inhibition of the generation of O
(C): effect of the four coumarins; (B) and (D): time-course of the CL-luc reaction in the presence of coumarin 1]. (E): Concentration-dependent inhibition of the PMA-
stimulated neutrophil CL-lum and CL-luc by ABAH. (F–K): Concentration-dependent increase of the generation of total ROS by neutrophils stimulated with SOZ (F–H) and
PMA (I–K), assessed by the luminol-enhanced chemiluminescence (CL-lum) assay [(F) and (I): effect of the four coumarins; (G, H, J and K): CL-lum produced by the
neutrophils treated with coumarins 1 and 2 (50 lM) in combination with ABAH (50 lM); (G) and (J): integrated CL-lum area; (H) and (K): time-course of the CL-lum reaction].
Results are expressed as mean ± standard deviation of at least four independent experiments, assayed in duplicate. Statistics: values not sharing the same letter (a–f) are
significantly different from each other (p < 0.05; ANOVA and Tukey’s post-hoc test).

The absorption spectra of coumarins 1 and 2 changed in the hydroxylated coumarins did not change in the absence of either
presence of MPO-H2O2: the intensity of the characteristic absorp- MPO or H2O2, suggesting that MPO metabolized these compounds
tion of coumarins at 324 nm decreased (Fig. 3E) in the first 5 min after this enzyme was activated by H2O2. The absorption spectra of
of reaction (Fig. 3D), whereas the typical absorption band of the coumarins 3 or 4 did not change significantly (Fig. 3C); therefore,
catalytic intermediates of MPO, compounds I and II, appeared in MPO-H2O2 did not oxidize these two coumarins under the assessed
the range of 410–430 nm. In addition, the spectra of these experimental conditions.
68 L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75

Fig. 3. Oxidation of the 7-hydroxycoumarin derivatives by myeloperoxidase (MPO). (A) Concentration-dependent enhancement of the MPO-H2O2-luminol (0.5 U mL1/
50 lM/20 lM) CL by the four coumarins, assayed in a reaction medium with 100 mM Cl-. (B) MPO-H2O2-luminol CL enhancement by coumarin 1, assayed in the presence of
100 mM Cl- or not. (C) Typical time-course of oxidation of the coumarins tested by MPO-H2O2; the absorption changes were recorded at 324 nm (coumarins 1 and 2) or
310 nm (coumarins 3 and 4) up to 20 min. (D) Oxidation of coumarin 1 (50 lM) by MPO-H2O2 (0.5 U mL1/50 lM) assayed in the presence of 100 mM Cl- or not; the
absorption changes were recorded at 324 nm. (E) Superimposed absorption spectra recorded during oxidation of coumarin 1 (50 lM) by MPO-H2O2 (0.5 U mL1/50 lM); the
dotted line is the spectra recorded before addition of MPO-H2O2. Data are expressed as mean ± standard deviation of at least three independent experiments, assayed in
duplicate.

In the cell-free systems, ABAH completely inhibited the oxida- The hyperfine coupling constant aH = 1.79 G of this signal agreed
tion of coumarin by MPO in PBS and in the chloride-free buffer, with the values reported in the literature [38]. The EPR signal in-
both in the presence and absence of luminol (data not shown). creased with rising concentration of coumarins 1 and 2 up to
90 lM, but decreased at higher concentrations (Fig. 4A.g); the
3.3. DPPH and HOCl scavenging effect intensity of this signal decreased along the reaction (data not
shown). Omission of HRP, H2O2, or the coumarins from the reaction
DPPH displayed reduced ability to inhibit the four coumarins mixture produced a low EPR signal relative to ascorbyl radical
(2.5–5.5%), and inhibition was not significantly different from the (Fig. 4A.a). HRP-H2O2 did not oxidize Asc at significant levels
control (3.68 ± 0.89%, at the highest concentration tested– (Fig. 4A.b) nor did the acetoxylated coumarins (3 or 4) or DMSO
100 lM). In contrast, coumarins 1–4 effectively scavenged HOCl: modify the EPR signal detected in this reaction significantly
their IC50 values were 4.15 ± 0.65, 4.34 ± 0.45, 7.05 ± 0.18, and (Fig. 4A.c,e and f).
11.56 ± 1.29 lM, respectively. The hydroxylated coumarins scav- The EPR signal of the ascorbyl radical and the absorption spec-
enged HOCl at least twice as effectively as their acetylated tra of the coumarins and Asc did not change significantly when
counterparts. these compounds were mixed in the absence of a peroxidase (data
not shown), indicating that the tested coumarins did not oxidize
3.4. Interaction between coumarins and ascorbic acid Asc directly. The H2O2 did not oxidize the coumarins or Asc di-
rectly under the assessed conditions.
A reaction mixture of Asc, HRP, H2O2 and one of the hydroxyl- Cyclic voltammetry revealed that Asc oxidized at
ated coumarins (1 or 2) gave a strong two-line EPR signal that +0.208 ± 0.019 V. Addition of equimolar concentrations of Asc
was characteristic of the ascorbyl free radical (Fig. 4A.d and e). and coumarin 1 or 2 to the buffer delayed the oxidation peak of
L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75 69

Fig. 4. Analysis of the interaction between ascorbic acid (Asc) and the tested coumarins (1–4). Data show representative results of two independent experiments.
Instrumental parameters are described in the Section 2. (A) EPR spectra of the ascorbyl radical detected in the reaction mixture containing HRP (20 UI/ml), H2O2 (0.5 mM), Asc
(0.4 mM), and coumarins 1–4 (90 lM) [(a): Asc alone; (b): Control (Asc + H2O2 + HRP); (c–f): Reaction (b) with DMSO and coumarins 1–4 (90 lM); (g): Integrated areas of the
ascorbyl EPR signals detected in the reaction mixture containing HRP-H2O2-Asc and the coumarins 1–4 at different concentrations]. (B) Cyclic voltammogram of Asc(0.5 mM)
and coumarin 1 alone or in combination [(a): Asc; (b): coumarin 1; (c): Asc + coumarin 1]. (C) Total charge of the coumarin peak calculated from cyclic voltammograms of
coumarin 1 alone (0.1–0.5 mM) or combined with 0.5 mM Asc. (D) Total charge of the Asc peak calculated from cyclic voltammograms of Asc alone (0.1–0.5 mM) or 0.5 mM
Asc combined with coumarin 1 (0.1–0.5 mM). Cyclic voltammetry (B–D) was performed in 0.1 M phosphate buffer pH 7.4, with glassy carbon working electrode at
100 mV s1 scan rate.

Asc to approximately +0.300 V, but did not alter the oxidation po- 3.6. EPR spin trapping
tential of coumarins 1 and 2: +0.699 ± 0.013 and +0.709 ± 0.008 V,
respectively (Fig. 4B). In the mixture, the total charge of coumarins We used DMPO and 4-POBN as spin traps to detect the couma-
increased in the presence of Asc (Fig. 4C), whilst the total charge of rin free radical intermediates produced by MPO. The peroxidase-
Asc decreased as a function of rising concentrations of the couma- catalyzed oxidation of the hydroxylated coumarins (1 or 2) in the
rins (Fig. 4D). Thus, these compounds act as antioxidants in this presence of DMPO gave the typical nine-line spectrum of 5,5-di-
cell-free system. methyl-1-pyrrolidone-2-oxyl-1 (DMPOX), whose characteristic
The anodic current peak height depended on the concentration hyperfine coupling constants aN = 7.2 G and aH = 4.1 G, appeared
of coumarins added to the electrochemical cell (data not shown) at g = 2.0085, agreeing with literature values [39–41] and the cor-
and its amplitude decreased after subsequent scanning. We did responding simulated spectra (Fig. 5A). The acetoxylated couma-
not detect any significant oxidation or reduction peaks for com- rins (3 or 4) did not produce a significant EPR signal in the
pounds 3 and 4, where the free hydroxyl group is acetylated (data presence of HRP-H2O2 and DMPO under the assessed conditions.
not shown). Luminol showed two oxidation peaks, at In the absence of coumarins or HRP, neither the EPR signal nor
+0.485 ± 0.011 and +0.618 ± 0.015 V. the absorption spectra (200–600 nm) modified significantly.
The intensity of the signal relative to DMPOX increased as a
function of the concentration of coumarin (1 and 2) from 10 to
120 lM, with the maximum intensity lying at 50 lM (Fig. 5A.i).
3.5. Physicochemical properties of the coumarins At this concentration, the EPR signal intensified with reaction time
until 5 min and decreased thereafter, suggesting that DMPOX
The physicochemical properties of coumarins 1–4 did not vary decomposed (Fig. 5A.j). Addition of superoxide dismutase to the
significantly. Their experimental partition coefficients were reaction mixture had negligible effect on the intensity of the EPR
1.46 ± 0.02, 1.79 ± 0.01, 1.07 ± 0.01, and 1.37 ± 0.02, respectively, signal (Fig. 5A.g), indicating that O
2 did not participate in the reac-
whereas the calculated partition coefficients were 1.61, 2.11, tions that produced DMPOX. Furthermore, oxidation of coumarin 1
1.17, and 1.67, respectively. The molecular volumes of coumarins by HRP-H2O2 did not generate significant amounts of O 2 , as as-
1–4 were 119.7, 134.2, 151.5, and 167.4 Å3, respectively.
70 L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75

Fig. 5. EPR spectra obtained from the oxidation of coumarins by HRP-H2O2 (20 U mL1/0.5 mM) using DMPO and 4-POBN as spin traps. Representative results of two
independent experiments. Instrumental parameters are described in the Section 2. (A) EPR spectra obtained with DMPO (80 mM) as spin trap after 5 min of reaction [(a):
Control (DMPO + H2O2 + HRP); (b–f): Reaction (a) with DMSO (0.8% v/v) and coumarins 1–4 (90 lM); (g): DMPO + H2O2 + HRP + coumarin 1 (90 lM) + superoxide dismutase
(SOD; 5000 U mL-1); (h): Computer simulation of the spectrum of DMPOX, where aN = 7.2 G, aH = 4.1 G, g = 2.0085, line width = 0.3 G; (i): Concentration-dependent increase
in the EPR signal area of DMPOX produced by DMPO + H2O2 + HRP + coumarin 1; (h): Time-course of the EPR signal area of DMPOX observed in the
DMPO + H2O2 + HRP + coumarin 1 (90 lM) reaction (c)]. (B) EPR spectra obtained with 4-POBN (100 mM) as spin trap after 15 min of reaction [(a): 4-POBN alone; (b) 4-
POBN + H2O2; (c): Control (4-POBN + H2O2 + HRP); (d–h): Reaction (c) with DMSO and coumarins 1–4 (90 lM); (i): Computer simulation of the spectrum of the 4-POBN/4-
POBN adduct, where aN = 14.9 G, a1H ¼ 1:8 G, and a2H ¼ 1:9 G; (j): Computer simulation of the spectrum of the POBN/coumarin adduct, where aN = 14.9 G, aH = 1.8 G; (k):
Time-course of EPR signal area observed in the 4-POBN + H2O2 + HRP + coumarin 1 (90 lM) reaction (e)].

sessed by the cell-free lucigenin-enhanced chemiluminescence a–d and g–h). This signal can be attributed to the adduct of 4-POBN
(data not shown). with its own radical 4-POBN, as previously described by McCor-
The oxidation of coumarins 1 or 2 by HRP-H2O2 in the presence mick et al. [42]. Computer simulations of the spectrum of such ad-
of 4-POBN as spin trap generated a characteristic twelve-line EPR duct using the determined parameters fit satisfactorily with the
spectrum (Fig. 5B.e,f). The signal intensified along time, reaching experimental data (Fig. 5B.i).
a maximum after 15 min (Fig. 5B.k). The corresponding hyperfine Coumarin radicals can also originate during the reaction, giving
coupling constants were a1N ¼ 14:90 G, a2N ¼ 1:85 G and rise to analogous POBN/coumarin adducts. However, these ad-
a1H ¼ 1:80 G. On the other hand, the HRP-H2O2-4-POBN reaction ducts would have a different pattern, showing a six-line spectrum,
produced a very weak EPR signal that did not change after addition considering that the coupling constants aN = 14.9 G and aH = 1.8 G
of DMSO or the coumarins to the medium, up to 20 min (Fig. 5B. are similar (Fig. 5B.j). These results suggest that the presence of
L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75 71

the hydroxylated coumarins increased the oxidation of 4-POBN by enzymatic activity: their inhibitory effect increased from
HRP-H2O2. 2.6 ± 2.5% to 5.7 ± 0.8%.
The hydroxylated coumarins inhibited the degranulation of
neutrophils three times more effectively than their acetylated ana-
3.7. Inhibition of elastase activity, degranulation and microbial killing logs (Fig. 6B). The inhibitory effect of coumarin 1 occurred at con-
centrations as low as 10 lM and was similar to the effect found at
We evaluated the ability of the four coumarins to inhibit the 100 lM. Cell treatment time did not increase this effect
elastase activity and suppress two important effector mechanisms significantly.
of neutrophils: degranulation and microbial killing. Coumarin 1 decreased the killing of C. albicans by neutrophils in
None of the tested coumarins inhibited the elastase activity of a concentration-dependent way (Fig. 6C). Untreated neutrophils
human neutrophils, at 100 lM, at least under the assessed condi- killed around 70% C. albicans added to the medium. At 10 lM,
tions (Fig. 6A). Longer incubation of elastase with the samples the mean neutrophil killing ability decreased to 60%; at 100 lM,
(from 20 to 60 min) did not significantly contribute to inhibiting this effector function of the neutrophil reduced to the same level
of MPO inhibition by ABAH (around 45%). In the absence of neutro-
phils, coumarin 1 and ABAH did not kill C. albicans, but HOCl at
200 lM killed 100% of the yeast added to the medium.

3.8. Cytotoxicity

Compared with the control, the four coumarins did not induce
significant LDH release or decrease the viability of neutrophils un-
der the assessed conditions (Table 1). After 60 min of treatment,
coumarin 1 did not induce apoptosis or necrosis; it did not reduce
cellular viability either. Thus, cytotoxicity does not underlie the
mechanism through which the tested coumarins inhibit the effec-
tor functions of neutrophils.

4. Discussion

We assessed whether 7-hydroxycoumarin (1) and its deriva-


tives (2–4) modulate the oxidative metabolism, elastase and mye-
loperoxidase activity, degranulation, and microbial killing ability of
human neutrophils. We also evaluated the physicochemical prop-
erties and antioxidant effect of the coumarins, as well as their po-
tential to scavenge HOCl in cell-free systems.
We evaluated the modulator effect of the tested coumarins on
the oxidative metabolism of SOZ- or PMA-stimulated human neu-
trophils by CL assays. We measured the production of O 2 and total
ROS by the lucigenin- and luminol-enhanced chemiluminescence
assays (CL-luc and CL-lum, respectively). SOZ is a model phagocytic
stimulus that binds to complement, IgG, and mannose membrane
receptors of neutrophils; it also triggers complex intracellular sig-
naling pathways [43]. Translocation and activation of protein ki-
nase C (PKC) is an important early event of the NADPH oxidase
assembly triggered by SOZ [43]. PMA is a soluble compound that
activates PKC directly [44].
All the tested coumarins inhibit the SOZ-stimulated CL-luc of
neutrophils more strongly than they inhibited the PMA-stimulated
CL-luc of neutrophils. Thus, inhibition of the activity of PKC by
these compounds mediates, in part, the decreased production of
2 by the neutrophils. The O2 scavenging by the hydroxylated
O 

coumarins [4,16] can also contribute to inhibiting CL-luc. Couma-


2 generation by human, rat, and rabbit
rins 1 and 2 inhibit the O
neutrophils [5,23] but not by murine RAW 264.7 macrophage cells
[45].
On the other hand, the four coumarins enhance the PMA- and
Fig. 6. Modulation of the effector functions of human neutrophils by the 7- SOZ-stimulated CL-lum of neutrophils. They exert a concentra-
hydroxycoumarin derivatives. (A) Inhibition of the elastase activity of neutrophils
tion-dependent effect on PMA-stimulated cells, but influence the
after 20 and 60 min of treatment with the coumarins. AAPV: methoxy-succinyl-Ala-
Ala-Pro-Val chloromethylketone, specific inhibitor of elastase activity: (B) Inhibi- SOZ-stimulated cells in a different way: as the concentration of
tion of the degranulation of neutrophils after 20 and 60 min of treatment with the the hydroxylated coumarins increases, the time to reach the max-
coumarins. (C) Inhibitory effect of coumarin 1 on the Candida albicans killing by imum CL-lum response and the peak height decrease. As a result,
neutrophils. HOCl was tested at 200 lM and ABAH (4-aminobenzoic acid hydra- the percentage of SOZ-stimulated CL-lum increases up 10 lM
zide) at 50 lM. Data are expressed as mean ± standard deviation of at least three
independent experiments assayed in duplicate. Statistics: values not sharing the
and diminishes at higher concentrations of these coumarins. Stim-
same letter (a–e) are significantly different from each other (p < 0.05; ANOVA and ulation of the neutrophils by SOZ releases much more MPO and
Tukey’s post-hoc test). less H2O2 than PMA [46]; hence, the amount of MPO released by
72 L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75

Table 1
Viability of neutrophils after treatment with 7-hydroxycoumarin derivatives.

Compounda 20 min 60 min


b c
Viable cells (%) LDH release (%) Viable cells (%)b LDH release (%)c Annexin-V+ cells (%)d IP+ cells (%)d
HBSS 98.00 ± 1.73 4.61 ± 1.62 77.10 ± 8.68 13.03 ± 4.33 0.13 ± 0.05 10.30 ± 2.45
DMSO 97.20 ± 0.84 4.19 ± 1.26 76.03 ± 4.79 14.62 ± 2.04 0.13 ± 0.05 9.89 ± 3.56
1 (10 lM) n.d. n.d. 78.15 ± 3.15 15.98 ± 3.67 0.13 ± 0.05 10.21 ± 2.24
1 (100 lM) n.d. n.d. 75.90 ± 5.51 13.70 ± 1.80 0.12 ± 0.04 8.47 ± 3.14
1 (200 lM) 98.13 ± 1.13 5.52 ± 1.73 n.d. n.d. n.d. n.d.
2 (200 lM) 97.88 ± 0.99 5.47 ± 1.24 n.d. n.d. n.d. n.d.
3 (200 lM) 98.50 ± 0.84 4.78 ± 3.46 n.d. n.d. n.d. n.d.
4 (200 lM) 97.50 ± 1.51 4.21 ± 2.01 n.d. n.d. n.d. n.d.

n.d.: Not determined.


a
HBSS: Hank’s Balanced Salt Solution (untreated cells); DMSO: dimethyl sulfoxide (0.1% v/v; vehicle control).
b
Determined by the Trypan Blue exclusion test by counting a total of 200 cells for each sample.
c
LDH release by the samples was compared with neutrophils completely lysed by Triton X-100.
d
Determined by flow cytometry analysis of 10,000 events.

the stimuli seems to influence the pattern of prooxidant effect of Addition of the 4-methyl group to 7-hydroxycoumarin does not
the coumarins. change its physicochemical properties – redox potential, partition
The CL-lum produced by the PMA- and SOZ-stimulated neutro- coefficient, and molecular volume – or its ability to modulate the
phils depends on the fusion of different types of intracellular effector functions of neutrophils investigated here significantly.
granules, which in turn is guided by the PKC and phosphatidylino- Recently, Witaicenis et al. [52] reported that 4-methyl-esculetin
sitol-3-kinase (PI3-K) signaling [43,44,47]. Together, our results suppressed the oxidative damage and inflammation in the intes-
suggest that the tested coumarins interfere in these pathways. tine of rats with acute colitis more effectively than esculetin
The fact that the coumarins significantly inhibit the degranulation (6,7-dihydroxycoumarin). Liver P-450 monoxygenases do not
of neutrophils at 10 lM supports our findings. These mechanisms metabolize the 4-methylcoumarins to toxic 3,4-epoxide intermedi-
reduced the CL-lum produced by SOZ-stimulated neutrophils ates, like many other coumarins [6,53]. This suggests that using
occurring at coumarin concentrations higher than 10 lM. Phagocy- 7-hydroxy-4-methylcoumarin instead of 7-hydroxycoumarin can
tosis of SOZ particles by neutrophils mobilizes the intracellular be advantageous.
granules more efficiently than PMA; thus, the interference of cou- The reason for the increase in CL-lum can be the reaction be-
marins in this process affects more strongly the SOZ- than the tween the hydroxylated coumarins with O 2 , yielding a free radical
PMA-stimulated effector functions of neutrophils. 7-Hydroxy- product that oxidizes luminol. The other hypothesis is that MPO
coumarin decreases the degranulation of mast cells [15] and inter- oxidizes the coumarins, which we investigated in the present
feres in early signaling mechanisms of J774 macrophages [21]. study. ABAH, a specific inhibitor of MPO, suppresses up to 60% of
Some monohydroxycoumarins inhibit the phosphorylation of the PMA- and SOZ-stimulated CL-lum of neutrophils. This com-
Akt/PKB in mouse RAW 264.7 macrophages [4]. pound significantly reduces the prooxidant effect of the coumarins,
Acetylation of the free hydroxyl group of coumarins 1 and 2 re- indicating that MPO oxidizes the coumarins to free radical prod-
duce their ability to modulate the PMA- and SOZ-stimulated neu- ucts which, in turn, oxidize luminol and increase the CL-lum. ABAH
trophil CL-luc and CL-lum, as well as their capacity to inhibit the completely inhibits the oxidative and peroxidative activities of
degranulation of neutrophils. This structural modification remark- MPO in cell-free systems, but suppresses only 80% of the HOCl gen-
ably decreases the prooxidant effect on the PMA-stimulated CL- eration by human neutrophils, without interfering in the processes
lum: the maximum CL-lum response of the acetylated compounds of degranulation and O 2 generation in these cells [46]. Moreover,
at 100 lM is around 80% lower than that of the hydroxylated ana- intracellular CL-lum originates via other reactions, such as luminol
logs. In addition, the acetylated coumarins are twofold more effec- oxidation by O 2 , HOCl, and HO [54].


tive to enhance the SOZ- than the PMA-stimulated CL-lum of In cell-free systems, MPO oxidizes the hydroxylated coumarins,
neutrophils. Together, these results suggest that the hydroxylated but not their acetylated counterparts. Thus, the free hydroxyl
coumarin is the pharmacologically active form, and the rate of group is essential to the prooxidant effect of these compounds.
hydrolysis of the acetylated drugs by intracellular esterases is The chloride ion probably competes with the coumarin and lumi-
higher in SOZ- than in PMA-stimulated neutrophils. nol in the active site of MPO, because the presence of this ion in
The stimulation of neutrophils with PMA and SOZ triggers the the assay buffer suppresses the CL-lum elicited by the coumarin.
generation of ROS in intracellular compartments much more effec- In the experimental system without luminol, MPO-H2O2 oxidizes
tively than in the extracellular milieu [47,48]. Therefore, the con- the coumarin more efficiently in the presence of Cl-. The HOCl
centrations of the four coumarins in the intracellular formed in this reaction also contributes to the oxidation of couma-
environment are sufficient for them to exert their pharmacological rin, because the four tested coumarins (hydroxylated and acety-
effects. Acetylation of the free hydroxyl does not seem to affect the lated) are efficient HOCl scavengers. The HOCl scavenging ability
intracellular bioavailability of the tested coumarins, because they of acetylated compounds was reported previously [4,31,55].
have similar partition coefficient and molecular volume. We previ- The antioxidant activity of coumarins depends on the type of
ously reported that this structural modification can increase or de- cell-free assay system. Hoult and Payá [5] reported that coumarins
crease the inhibitory effect of coumarin derivatives on the 2 , HOCl, alkylperoxyl radicals,
1 and 2 do not efficiently scavenge O
neutrophil oxidative metabolism, depending on the number and or inhibit lipid peroxidation. On the other hand, Kanimozhi et al.
position of the substituents [23,31,49]. In addition, esterification [16] reported that coumarin 1 effectively scavenge O 2 , HO , DPPH,


of hydroxyl groups has improved the pharmacological properties and ABTS+ (2,2-azino-bis-3-ethylbenzothiazoline-6-sulfonic acid
of catechin and quercetin derivatives in cell-mediated oxidative radical cation) radicals. Our experiments showed that compound
and inflammatory processes by favoring their interaction with 1 does not reduce DPPH significantly, in agreement with Lin
intracellular targets [50,51]. et al. [56]. Detecting this effect requires a longer reaction time
L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75 73

[16], due to the low reactivity of this coumarin. In addition, this capacity of neutrophils to kill C. albicans. We did not observe this
compound protects Asc from electrochemical oxidation but in- negative effect of the coumarins in macrophages, which are cells
creases the oxidation of Asc in the presence of a peroxidase. There- that contain low levels of MPO [22]. In fact, coumarin 1 improves
fore, the metabolization of the hydroxylated coumarins to the in vitro and in vivo phagocytic and microbial killing ability of
oxidizing species by peroxidases mediates the prooxidant effect murine monocytes and macrophages against Salmonella typhimuri-
of these coumarins toward Asc. Glutathione and Asc are the two um[22]. Compounds 1 and 2 do not kill C. albicans directly, and
major water-soluble intracellular antioxidants that act as redox they do not exhibit expressive antimicrobial activity against many
buffers by interacting with ROS, which is an important mechanism Gram-positive and Gram-negative bacteria [65,66].
of their protective function against oxidative stress [57,58]. In addition, inhibition of the effector functions of neutrophils is
Mammalian and plant peroxidases have been reported to not mediated by cytotoxicity of the four coumarins, at least under
produce ROS and aromatic oxyl radicals from several phenolic sub- the assessed conditions. In the same range of concentration used in
strates as part of their defense mechanism against pathogens [59]. this study, coumarin 1 is not toxic to J774 macrophages or to hu-
These enzymes also oxidize phenolphthalein, quercetin and other man and rabbit neutrophils [5,21,23].
phenolic compounds of plants [60–62]. Therefore, the presence of The prooxidant effect of hydroxycoumarins, at low levels, in-
hydroxyl groups makes the coumarins 1 and 2 likely candidates duces the apoptosis of U-937 cancer cells but not of normal cells
as substrates of endogenous peroxidases, such as MPO, lactoperox- [67]. The intracellular oxidative stress, at sub-lethal levels, can in-
idase, and eosinophil peroxidase. Miller et al. [63] reported that duce the synthesis of protective enzymes, such as glutathione syn-
HRP oxidizes 6-methoxy-7-hydroxycoumarin to transient free rad- thetase and AKR enzymes [17]. On the other hand, the antioxidant
ical intermediates. effect of these coumarins protects the host against oxidative injury.
Resonance can stabilize the free radical intermediates, or these 7-Hydroxycoumarin scavenges the O 2 , hydroxyl, and alkylperoxyl
radicals catalyze oxidative chain reactions. We found that the oxi- radicals [4,5,16], inhibits the production of NO by different macro-
dation of coumarin 1 by MPO generates highly reactive free radi- phage cell lines [19–21], and diminishes the secretion of O 2 and
cals that DMPO and 4-POBN may not be able to trap. This may H2O2 by macrophages obtained from healthy and Salmonella
happen because these free radicals oxidize the traps and prevent typhimurium-infected mice [22]. This coumarin improves the anti-
the formation of the corresponding coumarin-spin trap adduct. oxidant status of diabetic rats by enhancing both nonenzymic and
These radicals also oxidize Asc and luminol, so MPO oxidizes cou- enzymic antioxidants [11,18], and protects human lymphocytes
marin 1 faster than it oxidizes the other co-substrates added to the from radiation-induced generation of ROS [16] and human astrocy-
reaction medium, even if the concentration of this coumarin is toma 1321N1 cells against H2O2 and aldehyde toxicity [17]. It also
much lower than those of the other compounds. Interestingly, protects mice from neurotoxicity caused by exposure to 1-methyl-
the redox potential of Asc is lower than that of coumarin 1, which 4-phenyl-1,2,3,6-tetrahydropyridine, a compound that leads to the
could favor its oxidation by MPO or HRP; however, Asc is poorly production of ROS [68]. Moreover, the production of adequate
oxidized by HRP in the absence of the coumarin. amounts of ROS is important to regulate the course of acute and
Results obtained from EPR spin trapping experiments indicated chronic inflammatory responses and autoimmune inflammation
that HRP-H2O2 predominantly forms the 4-POBN/4-POBN adduct [3]. Hence, compounds that promote the formation of ROS can be
in the presence of hydroxylated coumarins. Other studies [42] have useful to treat rheumatoid arthritis and other inflammatory dis-
also observed this species, with the following spectroscopic param- eases [1,69].
eters: aH = 1.8 G, a1N ¼ 19:9 G, and a2N ¼ 1:85 G. Under our experi- Most of the biological effects of the coumarin derivatives re-
mental conditions, we did not find that HRP-H2O2 significantly ported herein take place in a range of concentrations considered
oxidized 4-POBN, suggesting that the presence of hydroxylated relevant for therapeutic purposes (1–10 lM) [53]. In physiological
coumarins is necessary to produce significant amounts of the buffer, the hydroxylated coumarin derivatives display significant

4-POBN radical. HRP-H2O2 likely oxidizes the hydroxylated MPO-mediated prooxidant effect at concentrations as low as
coumarins, and the resulting coumarin radical reduces 4-POBN to 1 lM. Recommended doses of coumarin range from 8 mg/day for
the 4-POBN radical, regenerating the parent phenolic compound the treatment of venous constriction to 7 g/day in antineoplastic
and establishing a redox cycling. A recycling mechanism may therapies [7,8]. After oral administration, the absorption rate of
explain the intensification of the EPR signal relative to the coumarin from the gastrointestinal tract can reach up to 83%,
4-POBN/4-POBN adduct, promoted by concentrations of coumarin and 60–75% of the dose is converted to the glucuronide of
1,000 times lower than the concentration of 4-POBN. Furthermore, 7-hydroxycoumarin [8]. It is not clear whether these glucuronide
the oxidation of DMPO to DMPOX seems to involve an analogous conjugates are pharmacologically active. However, mammalian
mechanism, because no EPR signal corresponding to the oxidized b-glucuronidases released from neutrophils at inflammatory
spin trap appears in the absence of the hydroxylated coumarins. sites may deconjugate 4-methylumbelliferyl-b-D-glucuronide and
Therefore, since the oxidation of the spin trap DMPO only occurs many flavonoid glucuronides and then deliver the free parent
in very strong oxidizing conditions, the peroxidase system drugs [70]. Therefore, it is possible that the 7-hydroxycoumarin
efficiently oxidizes the hydroxylated coumarins, yielding strong derivatives achieve therapeutic levels at inflammatory sites, but
oxidants [64]. further studies are needed to establish the best dosage form and
We also investigated how the pro- and anti-oxidant potential of administration route.
7-hydroxycoumarin (1) impacts the ability of the human neutro- In summary, the coumarins investigated here modulate the oxi-
phil to kill microbes. We found that this coumarin impairs the kill- dative metabolism and degranulation but not the elastase activity
ing of C. albicans, a phenomenon that is mediated by the decreased of human neutrophils. These compounds display antioxidant ef-
degranulation capacity of these cells and the reduced availability of fects that prevent neutrophils from generating O 2 , thereby scav-
O2 and other oxidizing species. Both the interference of this cou- enging HOCl and protecting Asc from oxidation. On the other
marin in the activity of MPO and its HOCl scavenging ability reduce hand, the hydroxylated compounds have prooxidant effects after
the availability of HOCl. Production of HOCl and other ROS is MPO-catalyzed reactions convert them to oxidizing species. These
important to kill Candida species, because some MPO-deficient species are highly reactive and oxidize Asc, the chemiluminescent
and chronic granulomatous disease patients suffer from frequent probe luminol, and the spin traps 4-POBN and DMPO. Considering
or severe infections with this microorganism [3]. Addition of the that the inflammatory site is a significant source of neutrophil
MPO inhibitor ABAH to the medium significantly decreases the MPO, formation of coumarin-derived reactive products is probably
74 L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75

part of the overall anti-inflammatory effect of 7-hydroxycoumarin [7] S.P. Felter, J.D. Vassalo, B.D. Carlton, G.P. Daston, A safety assessment of
coumarin taking into account species-specificity of toxicokinetics, Food Chem.
(1). Therefore, it is possible that both the anti- and prooxidant ef-
Toxicol. 44 (2006) 462–475.
fects of these compounds culminate in beneficial pharmacological [8] A. Lacy, R. O’Kennedy, Studies on coumarins and coumarin-related compounds
effects in vivo and contribute to suppressing the inflammatory re- to determine their therapeutic role in the treatment of cancer, Curr. Pharm.
sponse. 7-Hydroxycoumarin displays anti-inflammatory effect in Des. 10 (2004) 3797–3811.
[9] C.V.C. Ribeiro, M.A.C. Kaplan, Tendências evolutivas de famílias produtoras de
the murine model of allergic airway inflammation [10] and paw cumarinas em Angiospermae, Quím. Nova 25 (2002) 533–538.
edema [4,13–15]. However, the impaired C. albicans killing ability [10] J.F. Vasconcelos, M.M. Teixeira, J.M. Barbosa-Filho, M.F. Agra, X.P. Nunes,
of neutrophils is an important side-effect of 7-hydroxycoumarin A.M. Giulietti, R. Ribeiro-dos-Santos, M.B.P. Soares, Effects of umbelliferone
in a murine model of allergic airway inflammation, Eur. J. Pharmacol. 609
that needs to be considered during further research. In addition, (2009) 126–131.
the low cost and low in vitro and in vivo toxicity of these coumarins [11] B. Ramesh, K.V. Pugalendi, Antioxidant role of umbelliferone in STZ-diabetic
make them promising immunomodulating and anti-inflammatory rats, Life Sci. 79 (2006) 306–310.
[12] B. Ramesh, K.V. Pugalendi, Antihyperglycemic effect of umbelliferone in
drugs. streptozotocin-diabetic rats, J. Med. Food 9 (2006) 562–566.
[13] F.O. Lima, F.R. Nonato, R.D. Couto, J.M. Barbosa-Filho, X.P. Nunes, R.R. Santos,
5. Conclusion M.B.P. Soares, C.F. Villarreal, Mechanisms involved in the antinociceptive
effects of 7-hydroxycoumarin, J. Nat. Prod. 74 (2011) 596–602.
[14] T.J. Hardt, W.A. Ritschel, Investigation of the dose-response relationship upon
7-Hydroxycoumarin (1), 7-hydroxy-4-methylcoumarin (2), and intraperitoneal administration of coumarin and 7-hydroxycoumarin on the
their acetylated analogs (3 and 4, respectively) modulate the effec- carrageenan induced edema of the rats hind paw, Arzneim.-Forsch./Drug Res.
33 (1983) 1662–1666.
tor functions of human neutrophils studied: oxidative metabolism,
[15] D.O. Toyama, S. Marangoni, E.B.S. Diz-Filho, S.C.B. Oliveira, M.H. Toyama, Effect
degranulation and microbial killing. They act through different of umbelliferone (7-hydroxycoumarin, 7-HC) on the enzymatic, edematogenic
mechanisms, by interfering in intracellular signaling pathways, and necrotic activities of secretory phospholipase A2 (sPLA2) isolated from
scavenging ROS, interacting with MPO and generating oxidizing Crotalus durissus collilineatus venom, Toxicon 53 (2009) 417–426.
[16] G. Kanimozhi, N.R. Prasad, S. Ramachandran, K.V. Pugalendi, Umbelliferone
species. In general, the pro- and antioxidant effect of 7-hydroxy- modulates gamma-radiation induced reactive oxygen species generation and
coumarin in cellular and cell-free systems, as well as its ability to subsequent oxidative damage in human blood lymphocytes, Eur. J. Pharmacol.
modulate the effector functions of neutrophils here investigated, 672 (2011) 20–29.
[17] D. Li, E.M. Ellis, Inducible protection of human astrocytoma 1321N1 cells
are not significantly affected by adding the 4-methyl group but against hydrogen peroxide and aldehyde toxicity by 7-hydroxycoumarin is
are impaired by acetylating the free hydroxyl group. The balance associated with the upregulation of aldo-keto reductases, Neurotoxicology 33
between the anti- and prooxidant properties of these compounds (2012) 1368–1374.
[18] B. Ramesh, K.V. Pugalendi, Impact of umbelliferone on erythrocyte redox
is essential to achieve beneficial pharmacological effects without status in STZ-diabetic rats, Yale J. Biol. Med. 78 (2005) 131–138.
suppressing the microbial killing ability of neutrophils. Thus, it is [19] M. Kurokawa, W. Watanabe, T. Shimizu, R. Sawamura, K. Shirakic, Modulation
relevant to understand how these coumarins modulate the effector of cytokine production by 7-hydroxycoumarin in vitro and its efficacy against
influenza infection in mice, Antiviral Res. 85 (2010) 373–380.
functions of neutrophils, because these cells play important roles
[20] D. Zhao, M.N. Islam, B.R. Ahn, H.A. Jung, B.-W. Kim, J.S. Choi, In vitro
in the regulation of inflammatory processes. antioxidant and anti-inflammatory activities of Angelica decursiva, Arch.
Pharm. Res. 35 (2012) 179–192.
Acknowledgements [21] J.M. Timonen, R.M. Nieminen, O. Sareila, A. Goulas, L.J. Moilanen, M. Haukka, P.
Vainiotalo, E. Moilanen, P.H. Aulaskari, Synthesis and anti-inflammatory
effects of a series of novel 7-hydroxycoumarin derivatives, Eur. J. Med.
The authors thank Mr. Alcides S. Pereira and Mrs. Nadir Chem. 46 (2011) 3845–3850.
Mazzucato for technical assistance, and Prof. Dr. Gilberto U.L. [22] T. Stefanova, N. Nikolova, H. Neychev, G. Zlabinger, Phagocytosis and killing of
Salmonella by 7-hydroxycoumarin activated macrophages, Immunol. Invest.
Braga (FCFRP-USP, Brazil) for providing the C. albicans strain
41 (2012) 199–213.
used in the microbial killing assay. This study was supported [23] L.M. Kabeya, A. Kanashiro, A.E.C.S. Azzolini, F.M. Soriani, J.L.C. Lopes, Y.M.
by the Brazilian agencies: Fundação de Amparo à Pesquisa do Lucisano-Valim, Inhibitory effect of eight simple coumarins on the lucigenin
Estado de São Paulo (FAPESP, Grants 2000/06233-7, 2000/ enhanced chemiluminescence of rabbit neutrophils, Res. Commun. Mol.
Pathol. Pharmacol. 111 (2002) 103–114.
10641-3, 2002/09518-8, 2005/60596-8, 2007/02487-3, 2007/ [24] I.A. Vogel, A textbook of practical organic chemistry, Longmans, London, 1967.
00840-8), Coordenação de Aperfeiçoamento de Pessoal de Nível [25] J.L.C. Lopes, J.N.C. Lopes, J.C. Holanda, P.C. Vieira, Ação quimioprofilática de
Superior (CAPES), Conselho Nacional de Desenvolvimento alguns derivados cumarínicos sobre a esquistossomose, Rev. Fac. Farm. Odont.
Ribeirão Preto 13 (1976) 201–206.
Científico e Tecnológico (CNPq, grant 150302/2007-0), Instituto [26] F.S. Paula, L.M. Kabeya, A. Kanashiro, A.S.G. Figueiredo, A.E.C.S. Azzolini, S.A.
do Milênio Inovação e Desenvolvimento de Novos Fármacos e Uyemura, Y.M. Lucisano-Valim, Modulation of human neutrophil oxidative
Medicamentos (IM-INOFAR), Instituto do Milênio Redoxoma, metabolism and degranulation by extract of Tamarindus indica L. fruit pulp,
Food Chem. Toxicol. 47 (2009) 163–170.
Instituto Nacional de Ciência e Tecnologia (INCT) de Processos [27] A. Kanashiro, L.M. Kabeya, A.C.M. Polizello, N.P. Lopes, J.L.C. Lopes, Y.M.
Redox em Biomedicina, INCT de Inovação Farmacêutica and INCT Lucisano-Valim, Inhibitory activity of flavonoids from Lychnophora sp. on
de Bioanalítica. generation of reactive oxygen species by neutrophils upon stimulation by
immune complexes, Phytother. Res. 81 (2004) 61–65.
[28] A. Kanashiro, J.G. Souza, L.M. Kabeya, A.E.C.S. Azzolini, Y.M. Lucisano-Valim,
References Elastase release by stimulated neutrophils inhibited by flavonoids: importance
of the catechol group, Z. Naturforsch. [C] 62 (2007) 357–361.
[1] M. Hultqvist, L.M. Olsson, K.A. Gelderman, R. Holmdahl, The protective role of [29] T. Mosmann, Rapid colorimetric assay for cellular growth and survival:
ROS in autoimmune disease, Trends Immunol. 30 (2009) 201–208. application to proliferation and cytotoxicity assay, J. Immunol. Methods 65
[2] V. Kumar, A. Sharma, Neutrophils: Cinderella of innate immune system, Int. (1983) 55–63.
Immunopharmacol. 10 (2010) 1325–1334. [30] Y.M. Lucisano-Valim, L.M. Kabeya, A. Kanashiro, E.M.S. Russo-Carbolante,
[3] B. Amulic, C. Cazalet, G.L. Hayes, K.D. Metzler, A. Zychlinsky, Neutrophil A.C.M. Polizello, A.E.C.S. Azzolini, S.C. Silva, J.L.C. Lopes, C.A. Oliveira, B.
function: from mechanisms to disease, Annu. Rev. Immunol. 30 (2012) 459– Mantovani, A simple method to study the activity of natural compounds on the
489. chemiluminescence of neutrophils upon stimulation by immune complexes, J.
[4] L. Wu, X. Wang, W. Xu, F. Farzaneh, R. Xu, The structure and pharmacological Pharmacol. Toxicol. Methods 47 (2002) 53–58.
functions of coumarins and their derivatives, Curr. Med. Chem. 16 (2009) [31] M.F. Andrade, L.M. Kabeya, A.E.C.S. Azzolini, E.O.L. Santos, A.S.G. Figueiredo-
4236–4260. Rinhel, M.R.P. Paris, M.T. Pupo, F.S. Emery, Y.M. Lucisano-Valim, 3-
[5] J.R.S. Hoult, M. Payá, Pharmacological and biochemical actions of simple Phenylcoumarin derivatives selectively modulate different steps of reactive
coumarins: natural products with therapeutic potential, Gen. Pharmacol. 27 oxygen species production by immune complex-stimulated human
(1996) 713–722. neutrophils, Int. Immunopharmacol. 15 (2013) 387–394.
[6] B.G. Lake, Coumarin metabolism, toxicity and carcinogenicity: relevance for [32] M.S. Blois, Antioxidant determinations by use of a stable free radical, Nature
human risk assessment, Food Chem. Toxicol. 37 (1999) 423–453. 181 (1958) 1199–1200.
L.M. Kabeya et al. / Chemico-Biological Interactions 206 (2013) 63–75 75

[33] L.A. Marquez, H.B. Dunford, Mechanism of the oxidation of 3,5,30 ,50 - [52] A. Witaicenis, L.N. Seito, L.C. Di Stasi, Intestinal anti-inflammatory activity of
tetramethylbenzidine by myeloperoxidase determined by transient- and esculetin and 4-methylesculetin in the trinitrobenzenesulphonic acid model of
steady-state kinetics, Biochemistry 36 (1997) 9349–9355. rat colitis, Chem. Biol. Interact. 186 (2010) 211–218.
[34] R.R. Kitagawa, M.S.G. Raddi, N.M. Khalil, W. Vilegas, L.M. Fonseca, Effect of the [53] J.Z. Pedersen, C. Oliveira, S. Incerpi, V. Kumar, A.M. Fiore, P. De Vito, A.K. Prasad,
isocoumarin paepalantine on the luminol and lucigenin amplified S.V. Malhotra, V.S. Parmar, L. Saso, Antioxidant activity of 4-methylcoumarins,
chemiluminescence of rat neutrophils, Biol. Pharm. Bull. 26 (2003) 905–908. J. Pharm. Pharmacol. 59 (2007) 1721–1728.
[35] G.R. Buettner, L.W. Oberley, Considerations in the spin trapping of superoxide [54] K. Van Dyke, V. Castranova, Cellular chemiluminescence, vol. 1, CRC Press,
and hydroxyl radical in aqueous systems using 5,5-dimethyl-1-pyrroline-1- Boca Raton, 1987.
oxide, Biochem. Biophys. Res. Commun. 83 (1978) 69–74. [55] G. Morabito, D. Trombetta, K.S. Brajendra, K.P. Ashok, S.V. Parmar, C. Naccari, F.
[36] S. Pou, G.M. Rosen, Spin-trapping of superoxide by 5,5-dimethyl-1-pyrroline- Mancari, A. Saija, M. Cristani, O. Firuzi, L. Saso, Antioxidant properties of 4-
N-oxide: application to isolated perfused organs, Anal. Biochem. 190 (1990) methylcoumarins in in vitro cell-free systems, Biochimie 92 (2010) 1101–
321–325. 1107.
[37] A.T. do-Amaral, A.C. Oliveira, R. Neidlein, M. Gallaci, L. Caprara, Y. Miyazaki, [56] H.-S. Lin, S.-H. Tsai, C.-S. Chen, Y.-C. Chang, C.-M. Lee, Z.-Y. Lai, C.-M. Lin,
Physicochemical parameters involved in the lethal toxicity of N,N- Structure–activity relationship of coumarin derivatives on xanthine oxidase-
[(dimethylamino)ethyl]-4-substituted benzoate hydrochlorides: a QSAR inhibiting and free radical-scavenging activities, Biochem. Pharmacol. 75
study, Eur. J. Med. Chem. 32 (1997) 433–443. (2008) 1416–1425.
[38] G.O. Laroff, R.W. Fessenden, R.H. Schuler, The electron spin resonance spectra [57] Q. Chen, M.G. Espey, A.Y. Sun, J.-H. Lee, M.C. Krishna, E. Shacter, P.L. Choyke, C.
of radical intermediates in the oxidation of ascorbic acid and related Pooput, K.L. Kirk, G.R. Buettner, M. Levine, Ascorbate in pharmacologic
substances, J. Am. Chem. Soc. 94 (1972) 9062–9073. concentrations selectively generates ascorbate radical and hydrogen
[39] W. Adam, A. Kurz, C.R. Saha-Möller, DNA and 20 -deoxyguanosine damage in peroxide in extracellular fluid in vivo, Proc. Natl. Acad. Sci. 104 (2007) 8749–
the horseradish peroxidase-catalyzed autoxidation of aldehydes: the search 8754.
for the oxidizing species, Free Radic. Biol. Med. 26 (1999) 566–579. [58] S.L. Cuddihy, A. Parker, D.T. Harwood, M.C.M. Vissers, C.C. Winterbourn,
[40] M.M. Mossoba, I. Rosenthal, A.J. Carmichael, P. Riesz, Photochemistry of Ascorbate interacts with reduced glutathione to scavenge phenoxyl radicals in
porphyrins as studied by spin trapping and electron spin resonance, HL60 cells, Free Radic. Biol. Med. 44 (2008) 1637–1644.
Photochem. Photobiol. 39 (1984) 731–734. [59] N.C. Veitch, Horseradish peroxidase: a modern view of a classic enzyme,
[41] X. Shi, A. Lenhart, Y. Mao, ESR spin trapping investigation on peroxynitrite Phytochemistry 65 (2004) 249–259.
decomposition: no evidence for hydroxyl radical production, Biochem. [60] H. Yamasaki, S.C. Grace, EPR detection of phytophenoxyl radicals stabilized by
Biophys. Res. Commun. 203 (1994) 1515–1521. zinc ions: evidence for the redox coupling of plant phenolics with ascorbate in
[42] M.L. McCormick, G.R. Buettner, B.E. Britigan, The spin trap a-(4-pyridyl-1- the H2O2-peroxidase system, FEBS Lett. 422 (1998) 377–380.
oxide)-N-tert-butylnitrone stimulates peroxidase-mediated oxidation of [61] T. Miura, S. Muraoka, Y. Fujimoto, Inactivation of creatine kinase induced by
deferoxamine: implications for pharmacological use of spin-trapping agents, quercetin with horseradish peroxidase and hydrogen peroxide: pro-oxidative
J. Biol. Chem. 270 (1995) 29265–29269. and anti-oxidative actions of quercetin, Food Chem. Toxicol. 41 (2003) 759–
[43] S. Sergeant, L.C. McPhail, Opsonized zymosan stimulates the redistribution of 765.
protein kinase C isoforms in human neutrophils, J. Immunol. 159 (1997) 2877– [62] H.J. Sipe Jr., J.T. Corbett, R.P. Mason, In vitro free radical metabolism of
2885. phenolphthalein by peroxidases, Drug Metab. Dispos. 25 (1997) 468–480.
[44] P.M.-P. Dang, S. Rais, J. Hakim, A. Périanin, Redistribution of protein kinase C [63] R.W. Miller, J.-C. Sirois, H. Morita, The reaction of coumarins with horseradish
isoforms in human neutrophils stimulated by formyl peptides and phorbol peroxidase, Plant Physiol. 55 (1975) 35–41.
myristate acetate, Biochem. Biophys. Res. Commun. 212 (1995) 664–672. [64] G.D. Mao, P.D. Thomas, M.J. Poznansky, Oxidation of spin trap 5,5-dimethyl-1-
[45] A. Murakami, Y. Nakamura, T. Tanaka, K. Kawabata, D. Takahashi, K. pyrroline-1-oxide in an electron paramagnetic resonance study of the reaction
Koshimizu, H. Ohigashi, Suppression by citrus auraptene of phorbol ester- of methemoglobin with hydrogen peroxide, Free Radic. Biol. Med. 16 (1994)
and endotoxin-induced inflammatory responses: role of attenuation of 493–500.
leukocyte activation, Carcinogenesis 21 (2000) 1843–1850. [65] M. Kawase, B. Varu, A. Shah, N. Motohashi, S. Tani, S. Saito, S. Mahapatra, S.G.
[46] A.J. Kettle, C.A. Gedye, M.B. Hampton, C.C. Winterbourn, Inhibition of Dastidar, A.N. Chakrabarty, Antimicrobial activity of new coumarin
myeloperoxidase by benzoic acid hydrazides, Biochem. J. 308 (1995) 559–563. derivatives, Arzneim. Forsch. – Drug Res. 51 (2001) 67–71.
[47] J. Bylund, K.L. Brown, C. Movitz, C. Dahlgren, A. Karlsson, Intracellular [66] J.T. Konc, E. Hejchman, H. Kruszewska, I. Wolska, D. Maciejewska, Synthesis
generation of superoxide by the phagocyte NADPH oxidase: how, where, and and pharmacological activity of O-aminoalkyl derivatives of 7-
what for?, Free Radic Biol. Med. 49 (2010) 1834–1845. hydroxycoumarin, Eur. J. Med. Chem. 46 (2011) 2252–2263.
[48] C. Dahlgren, A. Karlsson, Respiratory burst in human neutrophils, J. Immunol. [67] M.E. Riveiro, A. Moglioni, R. Vazquez, N. Gomez, G. Facorro, L. Piehl, E.R. De
Methods 232 (1999) 3–14. Celis, C. Shayo, C. Davio, Structural insights into hydroxycoumarin-induced
[49] L.M. Kabeya, C.H.T.P. da Silva, A. Kanashiro, J.M. Campos, A.E.C.S. Azzolini, apoptosis in U-937 cells, Bioorg. Med. Chem. 16 (2008) 2665–2675.
A.C.M. Polizello, M.T. Pupo, Y.M. Lucisano-Valim, Inhibition of immune [68] S.R. Subramaniam, E.M. Ellis, Neuroprotective effects of umbelliferone and
complex-mediated neutrophil oxidative metabolism: a pharmacophore esculetin in a mouse model of Parkinson’s disease, J. Neurosci. Res. 91 (2013)
model for 3-phenylcoumarin derivatives using GRIND-based 3D-QSAR and 453–461.
2D-QSAR procedures, Eur. J. Med. Chem. 43 (2008) 996–1007. [69] M. Hultqvist, P. Olofsson, K.A. Gelderman, J. Holmberg, R. Holmdahl, A new
[50] Y.C. Chen, S.C. Shen, W.R. Lee, W.C. Hou, L.L. Yang, T.J.F. Lee, Inhibition of nitric arthritis therapy with oxidative burst inducers, PLoS Med. 3 (2006) 1625–
oxide synthase inhibitors and lipopolysaccharide induced inducible NOS and 1636.
cyclooxygenase-2 gene expressions by rutin, quercetin and quercetin [70] R. Bartholomé, G. Haenen, P.C.H. Hollman, A. Bast, P.C. Dagnelie, D. Roos, J.
pentaacetate in RAW264.7 macrophages, J. Cell Biochem. 82 (2001) 537–548. Keijer, P.A. Kroon, P.W. Needs, I.C.W. Arts, Deconjugation kinetics of
[51] M.L. Vuotto, R. Miranda, A. Ritieni, A. Basile, L. Ricciardi, R. Di Prisco, G. glucuronidated phase II flavonoid metabolites by b-glucuronidase from
Nicolosi, N. Mascolo, Improvement of (+)-catechin inhibitory activity on neutrophils, Drug Metab. Pharmacokinet. 25 (2010) 379–387.
human PMN respiratory burst by (+)-3-O-propionyl and ()-3-O-valeryl
substitution, J. Pharm. Pharmacol. 55 (2003) 399–405.

You might also like