You are on page 1of 10

Parasitology International 67 (2018) 627–636

Contents lists available at ScienceDirect

Parasitology International
journal homepage: www.elsevier.com/locate/parint

Chemoprevention of Leishmaniasis: In-vitro antiparasitic activity of T


dibenzalacetone, a synthetic curcumin analog leads to apoptotic cell death
in Leishmania donovani☆
Indira Singh Chauhana, G. Subba Raob, Jai Shankarc, Lalit Kumar Singh Chauhanc,

Govind J. Kapadiad,1, Neeloo Singha,
a
Biochemistry Division, CSIR Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
b
Global Biotechnology Resource Center, 145 Rosewood Drive, Streamwood, IL 60107, USA
c
Transmission Electron Microscopy, CSIR Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow 226001, India
d
Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington, DC 20059, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Curcumin is the major phenolic compound found in turmeric, a dry powder of rhizomes and roots of the plant,
Leishmania donovani Curcuma longa L., which is widely used as spice and food colorant around the world, and in herbal medicinal
A curcumin analog dibenzalacetone (DBA) practice in Asian countries. The present study reports the leishmanicidal activity of trans-dibenzalacetone (DBA),
Apoptosis a synthetic monoketone analog of curcumin, against Leishmania donovani parasites. We for the first time report
the antiproliferative effect of a curcumin analog (DBA) on the intracellular amastigotes of L. donovani, the
clinically more relevant stage of the parasite than its promastigotes stage. The leishmanicidal effect of DBA was
further confirmed by scanning and transmission electron microscopies. Cell growth was arrested in G0/G1 phase
with increased concentration of cytosolic calcium and dissipation of mitochondrial membrane potential. Further,
the unique trypanothione/trypanothione reductase (TR) system of Leishmania cells was significantly inhibited
by DBA. This economically synthesizable simple monoketone analog of curcumin has the potential for field use
against visceral leishmaniasis which is currently widespread in tropical and subtropical developing countries of
the world. In conclusion, we have identified an analog of curcumin for potential applications against leishma-
niasis, based on its strong antiparasitic activity and low toxicity. This curcumin analog compares favorably, at
least in vitro, with the existing medication miltefosine.

1. Introduction leishmaniasis is caused by a protozoan parasite which they named


Leishmania donovani [5]. Being an opportunistic infection with up to
Trypanosomatids are a family of kinetoplastid protozoa comprised 70% of adult leishmaniasis related to HIV infection, it is the second-
of flagellated protists characterized by the presence of a single fla- largest parasitic disease in the world (after malaria) with 500,000 new
gellum [1]. Leishmaniasis is one of the three major human parasitic cases of VL reported annually [6]. VL has been classified by WHO as one
diseases caused by trypanosomatids, the other two being sleeping of the most neglected tropical diseases which impacts mostly the
sickness caused by Trypanosoma brucei and Chagas disease caused by poorest populations and for which no adequate therapy currently exists
T. cruzi. Leishmania donovani is transmitted to humans by sandflies and [7]. The orally effective drugs, such as miltefosine, a phosphocholine
responsible for visceral leishmaniasis (VL), the fatal form if untreated, derivative and sitamaquine, an aminoquinoline have found limited
in 88 tropical and subtropical countries in every continent except success in India, Brazil and Kenya, all exhibit serious toxic side-effects
Australia and Antarctica [2, 3]. There is evidence for its prevalence and develop resistance [8–11]. Recent trend has been to employ com-
among early Egyptians dating back to 3500 BC by recent detection of L. bination therapy, such as antimonials or miltefosine with antileismanial
donovani DNA in the bone tissue samples from ancient mummies of the antibiotics to overcome drug resistance [12]. However, most of these
era [4]. In 1903, Leishman and Donovan were the first to report that therapies are not practical for field use in treating VL in economically-


This article is dedicated to Prof. Govind J. Kapadia

Corresponding author.
E-mail address: neeloo888@yahoo.com (N. Singh).
1
Deceased.

https://doi.org/10.1016/j.parint.2018.06.004
Received 2 February 2018; Received in revised form 27 April 2018; Accepted 8 June 2018
Available online 18 June 2018
1383-5769/ © 2018 Elsevier B.V. All rights reserved.
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

challenged developing countries. Thus, there is urgent need for new 2. Materials and methods
orally effective, affordable drugs without toxicity and drug resistance in
managing VL infection and improving quality of life of large popula- 2.1. Synthesis of trans-Dibenzalacetone (DBA)
tions afflicted with this debilitating parasitic disease in both developing
and developed countries of the world. DBA was synthesized as described earlier [37, 38] by the one-step
Curcumin [(1E,6E)-1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-hepta- Aldol condensation reaction of benzaldehyde in ethanol with acetone
diene-3,5-dione] is the principal phenolic compound responsible for the under basic conditions utilizing aqueous sodium hydroxide solution at
yellow color of turmeric, a dry powder of rhizomes and roots of the room temperature with a yield of > 85%. Recrystallization from aqu-
perennial plant, Curcuma longa L. belonging to the ginger family, eous alcohol twice yielded DBA as bright yellow crystals, melting point
Zingiberaceae [13]. Over the centuries, turmeric has been used ex- 103–104 °C and its purity established by thin-layer chromatography, as
tensively as a spice; food colorant and fabric dye, and in herbal medical described earlier [39]. The chemical structure of DBA was confirmed by
practice in India, China and other Asian countries [14]. The European infrared (strong signal for the presence of carbonyl group at
Union and the US Food and Drug Administration consider curcumin, 1649 cm−1with none attributable to hydroxyl group), ultraviolet (λmax
designated as yellow food color E100, safe for human consumption 326 nm, log εmax4.53 for conjugated carbonyl compound), 1H-nuclear
(http://www.feingold.org/Research/PDFstudies/colors.pdf). It is a magnetic resonance (vinyl protons doublet of doublets, 16 Hz at 7.74
strong antioxidant exhibiting remarkable pharmacological properties, and 7.09 ppm, each for 2H for the benzylic protons and for the protons
such as anti-inflammatory, antimicrobial and cancer chemoprevention α to the carbonyl carbon, respectively) and mass spectra (molecular
through a variety of mechanisms involving multiple targets [15–17]. In ion, M+ at m/z 234, M+–H ion at m/z 233 and ion at m/z 131 from
clinical trials, curcumin exhibited no adverse effects in humans at high cleavage at carbonyl carbon leading to ion at m/z 103 from loss of CO)
doses (8-12 g/day) [16, 18]. However, instability, poor cellular uptake [39, 40].
and bioavailability of curcumin and its naturally occurring analogs,
curcumin II (demethoxycurcumin) and curcumin III (bisdemethox- 2.2. Parasite culture and measurement of cell viability
ycurcumin) have hindered their development into clinically useful
drugs to treat systemic leishmaniasis [19]. Since antioxidant capacity of Promastigotes and intracellular amastigotes of L. donovani strain
curcumin and its analogs with β-diketone [20–23] or monoketone [24, (MHOM/IN/80/DD8) used in this study were routinely cultured as
25] moiety in their structure is considered essential for their biological described previously [41]. Stock solution of DBA (1 mg/ml) was made
activities, numerous curcumin analogs with improved stability, anti- in 0.1% (v/v) of DMSO and stored in aliquots at 4 °C. The drug was
oxidant capacity, cell penetration and bioavailability profile have been diluted in culture media immediately prior to each experiment to pro-
synthesized for structure-antioxidant/biological activity studies [19, vide series of drug concentrations (0, 10, 20, 40, 80 and 160 μg/ml), as
25–27]. Previous investigations have demonstrated antiparasitic activ- needed.
ities of curcumin [21, 28–30] and related synthetic curcuminoids [31, Resazurindye/AlamarBlue® (7-hydroxy-3H-phenoxazin-3-one-10-
32]. oxide) (Invitrogen, Cat. No. DAL1025, Carisbad, CA) was employed for
The average daily intake of turmeric in India is estimated to be measuring parasite (promastigotes) viability. Logarithmic phase pro-
38–190 mg (equivalent to 0.38–9.5 mg/day of curcumin) where it is mastigotes of L. donovani (50,000 cells/200 μl/well) were seeded in 96-
widely used in popular curry dishes for its unique flavor and signature well microtiter plates (Greiner, Bio-one, Germany) in the presence of
bright yellow color [33, 34]. In some rural areas of this country, its increasing concentrations (0–40 μg/ml) of DBA, incubated at 25 °C for
consumption is as high as 3.8 g/day. Effect of curcumin on systemic 24 h. Miltefosine was used as the standard drug. AlamarBlue® (20 μl)
leishmaniasis appears to be dose dependent: in high doses it has anti- was added to each well and the plate was further incubated at 25 °C for
parasitic effect and in low doses it increases parasite load in critical 4 h. Absorbance was measured in a microplatereader (Biotek, Epoch)
organs and exacerbates VL [34, 35]. Formation of reactive oxygen using λ = 570 nm as test wavelength (resorufin) and λ = 600 nm as
species (ROS) and imbalance of calcium homeostasis account for high reference wavelength (resazurin) serving as blank (normalized to the
doses of curcumin inducing cell death of L. donovani [21]. In contrast, 600 nm value). The percent of cell-viability of DBA treated parasites
curcumin in low doses suppresses interferon γ and nitric oxide pro- was analyzed by following equation: [untreated control λ
duction by activation of the anti-inflammatory protein called PPARγ (570–600 nm) – treated set λ (570–600 nm)]/untreated control λ
which results in suppression of the body's initial acute inflammatory (570–600 nm) × 100.
immune response against invading parasite and helps them evade the
host immune attack [35]. Thus, currently there is an inconclusive re- 2.3. In vitro evaluation of antileishmanial activity of DBA in intracellular
search on the effect of dietary intake of curcumin in systemically amastigotes
Leishmania infected population in India, especially those in rural areas.
However, topical ointments and skin injections of turmeric and cur- The BALB/c mouse macrophage cell line J774A.1 was infected with
cumin have proven to be helpful in the treatment and prevention of stationary phase promastigotes of L. donovani as previously described
cutaneous leishmaniasis [36]. [41]. Infected macrophages were treated with increasing concentra-
The present study describes the leishmanicidal activity of (1E, 4E)- tions (0–20 μg/ml) of DBA, incubated at 37 °C in 5% CO2 for 24 h. After
1,5-diphenyl-1,4-pentadien-3-one (trans-dibenzalacetone, DBA), a indicated incubation time, infected macrophages treated with or
simple, synthetic monoketone analog of curcumin, against promasti- without DBA were fixed with ice-cold methanol on slides which were
gotes and intracellular amastigotes of L. donovani. The potential me- then submerged in 10% (v/v) Giemsa staining solution (Thomas Baker)
chanism of leishmanicidal activity of DBA was studied by cell cycle for 45 min, followed by water rinse and let dry. The slides were viewed
progression and ultrastructure of treated parasites. These leishmani- on an inverted bright field microscope (IX73 Inverted Microscope,
cidal results are of particular interest since DBA is a simple monoketone Olympus). At least 100 macrophages per well were counted from du-
curcumin analog easily synthesized in high yield (> 85%) from in- plicate cultures and the percentage of infected macrophages were cal-
expensive ingredients by a one-step process at room temperature and culated using the formula [42]: % reduction = number of amastigotes
has potential for economically treating VL currently wide-spread among per 100 macrophages (treated samples)/number of amastigotes per 100
poor populations of the world. macrophages (infected control) × 100.
In vitro antileishmanial activity was expressed as the concentration
inhibiting parasite growth by 50% (IC50) and was analyzed by plotting
percentage of cell viability versus log growth concentration of DBA.

628
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

(caption on next page)

629
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

Fig. 1. [A] Cell viability assessment by AlamarBlue® (7-Hydroxy-3H-phenoxazin-3-one-10-oxide) shows reduction in viability of promastigotes treated with different
concentrations (0, 10, 20 and 40 μg/ml) of DBA for 24 h. (a) Graph shows percentage of cell viability against log value of DBA concentrations (μg/ml). (b) Changes of
medium color associated with AlamarBlue®reduction from blue (oxidized) to pink (reduced) in triplicate wells. [1] Control (cells without DBA) shows pink color and
[2 and 3] promastigotes treated with different concentrations of DBA show blue color where a blue color in the well was interpreted as no cell growth, and pink color
was scored as growth. The data are presented as mean ± standard deviation of three independent experiments. [B] In vitro analysis of antileishmanial activity of
DBA in intracellular amastigotes: Macrophages (4000 cells/well, final volume 200 μl) were infected with promastigotes of L. donovani in a ratio of 8:1 (parasites/
macrophages) and infected macrophages were treated with increasing concentrations (0, 5, 10, 15 and 20 μg/ml) of DBA for 24 h. After 24 h, treated or untreated
cells were stained with Giemsa stain and the slides were viewed on an inverted bright field microscope (IX73 Inverted Microscope, Olympus). The 50% inhibitory
concentration (IC50) was obtained by plotting the graph of percentage of cell viability against log value of DBA concentrations (μg/ml). The results were taken as the
mean of duplicate experiments and p < .05 has been considered as the level of significance. [C] Chemical structure of (a) curcumin [(1E,6E)-1,7-bis(4-hydroxy-3-
methoxyphenyl)-1,6-heptadiene-3,5-dione] (b) a curcumin analog, trans-dibenzalacetone (DBA) [(1E,4E)-1,5-diphenyl-1,4-pentadien-3-one]. [D](a) Cytotoxicity in
macrophages: Macrophages (50,000 cells/well, final volume 200 μl) were treated with increasing concentrations (0, 10, 20, 40, 80 and 160 μg/ml) of DBA for 24 h.
After 24 h, the viability of the macrophages was estimated by AlamarBlue® cell viability reagent. The 50% cytotoxic concentration (CC50) was determined by
logarithmic regression analysis using GraphPrism 5 software. (b) Growth in the medium is indicated by change in color from blue to pink. Results are presented as
mean ± SD; n = 3 and p < .05 has been considered as the level of significance. [E] PI uptake analysis: histograms show that treatment of Leishmania parasites with
DBA leads to (PI-positive cells M2) cell death in a concentration-dependent manner [a, b, c and d represent 0, 10, 20 and 40 μg/ml respectively]. All data depicted in
this figure are representative of at least three replicates. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of
this article.)

2.4. In vitro cytotoxicity against BALB/c mouse macrophages 2.7. Ultrastructural analysis by transmission electron microscopy (TEM)

The BALB/c mouse macrophage cell line J774A.1 was seeded in 96- Logarithmic-phase L. donovani promastigotes (2 × 107 cells/ml)
well culture plates (104 cells/200 μl/well). The plates were incubated were incubated with IC50 dose (17.80 μg/ml) of DBA for 24 h at 25 °C in
overnight in a CO2 incubator in an atmosphere of 95% air and 5% M199 medium. The parasites were then washed with PBS (pH 7.2) prior
CO2at 37 °C. DBA in different concentrations (0–160 μg/ml) was dis- to fixing in 2.5% glutraldehyde in sodium cacodylate buffer (pH 7.2) for
pensed in triplicate and the remaining three wells were used as con- 2 h at 4 °C. Fixed parasites were centrifuged at 3500 x g for 10 min and
trols. After 24 h, 20 μl of AlamarBlue® was added to each well and the washed 3 times with 0.1 M sodium cacodylate buffer and post-fixed in
plate was further incubated at 37 °C for 4 h. Absorbance was measured 1% osmium tetraoxide for 2 h. Post-fixed parasites were further washed
in a microplate reader as described before [43]. The 50% cytotoxic with sodium cacodylate and dehydrated in ascending acetone series
concentration (CC50) for the above mentioned assays was determined (15, 30, 60 and 100%), embedded in araldite-DDSA mixture and baked
by logarithmic regression analysis using GraphPrism 5 software. at 60 °C for 48 h. The baked blocks were cut (60–80 nm thick) by an
ultramicrotome (Leica EM UC7, Vienna, Austria), mounted on copper
grids, and double stained with uranyl acetate and lead citrate. The
2.5. Propidium iodide (PI) uptake assay stained sections were examined by TEM (TECNAI G2 SPIRIT, FEI,
Netherland) equipped with GatanOrius camera at 80 KV [41].
Logarithmic phase promastigotes of L. donovani (106 cells/2 ml/
well) were seeded in 6-well microtiter plates in the presence of different 2.8. Cell cycle analysis
concentrations (0–40 μg/ml) of DBA and incubated at 25 °C for 24 h.
The cells were then centrifuged (3500 x g for 10 min), washed once In order to determine the changes induced by DBA on the cell cycle,
with PBS (pH 7.2), re-suspended in 50 μg/ml final concentration of PI 2 × 106 cells/ml of log phase L. donovani promastigotes were treated
(Sigma-Aldrich), and incubated for 30 min in the dark at room tem- with different concentrations (0–40 μg/ml) of DBA for 24 h and were
perature. Unbound PI was removed by washing and samples were fixed by incubation in 70% ethanol and 30% PBS for 1 h at 4 °C. Fixed
processed on a FACSCalibur flow cytometer (BD Bioscience, San Jose, cells were washed in 1 ml PBS and re-suspended in 1 ml PBS with
CA, USA) and analyzed using CellQuest Pro software. Twenty thousand 100 μg/ml of RNAse A (Cat. No.EN0531, Fermentas) and PI (10 μg/ml).
events from each sample were acquired to ensure adequate data and at The cells were then incubated at 35 °C for 45 min and analyzed using
least three independent experiments were carried out [44, 45]. FACSCalibur flow cytometer. Cell cycle distribution was modeled using
ModFit LT for Mac V3.0. Ten thousand events from each sample were
acquired to ensure adequate data and at least three independent ex-
2.6. Analysis of topological alterations by scanning electron microscopy periments were run [46].
(SEM)

2.9. Analysis of externalized phosphatidylserine (PS)


Logarithmic phase promastigotes of L. donovani were incubated in
the presence of DBA washed with PBS (pH 7.2) prior to fixing in 2.5%
Double staining with Annexin-V and PI was used to measure the
glutraldehyde in sodium cacodylate (Ladd Research Industries, USA)
apoptotic effects of DBA on the plasma membrane of L. promastigotes
buffer (pH 7.2) for 4 h at 4 °C. Fixed samples were washed with sodium
(Annexin V-FITC Apoptosis Detection kit, Sigma). The log phase L.
cacodylate buffer (0.1 M) three times and the suspensions were placed
donovani promastigotes (2 × 106 cells/ml) were treated with different
on poly-L-lysine-coated glass slides and allowed to adhere for 15 min at
concentrations (0–40 μg/ml) of DBA for 24 h and treated cells were
room temperature. The samples were post-fixed in 1% osmium tetra-
analyzed on a FACSCalibur flow cytometer and 10,000 events from
oxide (OsO4) in dark at 4 °C for 4 h, washed and dehydrated in ethanol
each sample were acquired to ensure adequate data and at least three
series from 15 to 100%. Dehydrated samples were critical point dried
independent experiments were conducted [41].
by K850 Critical Point Dryer (Quorum Technologies Ltd., UK) and taken
on stubs for coating by gold sputter coater (SC7620, mini sputter coater,
Quorum 1040 Technologies Ltd., UK). DBA treated parasites were ex- 2.10. Terminal Deoxynucleotidyltranferase(TdT)-mediated dUTP -nick end
amined under scanning electron microscope (Quorum FEG 450, FEI, labeling (TUNEL) assay
Netherland).
DNA fragmentation within L. donovani promastigotes was analyzed
by Terminal Deoxynucleotidyltranferase(TdT)-mediated dUTP Nick

630
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

Fig. 2. Scanning electron microscopy (SEM) shows morphological alterations in (b) promastigotesof L. donovani treated with the IC50value (17.80 μg/ml) of DBA for
24 h (a) as compared to untreated cells. Scale bars 5 μm and 10 μm.

End Labeling (TUNEL) using an APO-BRDU flow cytometry assay kit (0–40 μg/ml) of DBA and incubated at 25 °C for 24 h. Then, the cells
(Sigma)as described previously [41]. Promastigotes were incubated were centrifuged (3500 x g for 5 min), washed once with PBS (pH 7.2),
with different concentrations (0–40 μg/ml) of DBA at 25 °C for 24 h, re-suspended in 5 μl of Fluo-4 AM (1 mM) and incubated for 30 min in
and treated cells were analyzed for fluorescence on a FACSCalibur flow the dark at room temperature. After washing, samples were processed
cytometer equipped with dual pass FITC/PI filter set. CellQuest Pro on a FACSCalibur flow cytometer and analyzed using CellQuest Pro
software was used for flow cytometer data acquisition and analysis software. Ten thousand events from each sample were acquired to en-
which represented three independent experiments with 10,000 events sure adequate data and at least three independent experiments were
from each sample to ensure adequate data. run.

2.11. Monitoring of changes in intracellular Ca2+ levels 2.12. Measurement of mitochondrial membrane potential (Δψm)

Changes in intracellular calcium levels were measured by the Mitochondrial damage upon treatment with DBA in L. donovani
fluorometric dye fluo-4 acetoxymethyl ester (Fluo 4-AM) (Cat. No·F- promastigotes was assessed by flow cytometry using a cell-permeable
14201, Molecular Probes) [47, 48]. Logarithmic phase promastigotes of dye, MitoTracker deep red (Molecular Probes, USA) [49]. L. donovani
L. donovani (1 × 106 cells, final volume 2 ml/well) were seeded in 6- promastigotes were treated with different concentrations (0–40 μg/ml)
well microtiter plates in the presence of different concentrations of DBA for 24 h, washed in PBS and loaded in dark for 30 min with

631
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

Fig. 3. Evaluation of ultrastructural damage by


transmission electron microscopy (TEM):
Promastigotes of L. donovani were incubated with the
IC50 value (17.80 μg/ml) of DBA for 24 h: [A] control
(untreated); [B] DBA treated cells, n: nucleus; k: ki-
netoplast; m: mitrochondria; pf: pocket flagellar; pm:
plasma membrane; G: golgi body; g: glycosome; a:
acidocalcisomes; ER: endoplasmic reticulum; sm:
sub-membrane microtubules; v: contractile vacuole.
Scale bars 1.0 μm and 0.5 μm.

MitoTracker (10 μM) as per manufacturer's instructions. Analysis for 3. Results


mean fluorescence intensity (MFI) was done using a FACSCalibur flow
cytometer and CellQuestPro software. Ten thousand events from each 3.1. In vitro activity of DBA against L. donovani and mammalian
sample were acquired to ensure adequate data and at least three in- cytotoxicity
dependent experiments were run.
The antileishmanial activity of DBA was tested initially against
promastigotes of L. donovani. Promastigotes which resulted in the loss
2.13. Measurement of intracellular non-protein thiols
of parasite viability in a concentration-dependent manner (Fig. 1A).
The DMSO solvent employed had no effect on the parasite viability. The
The probe, 5-chloromethylfluorescein-diacetate (CMFDA,
IC50 value of DBA for intracellular amastigotes (7.43 ± 1.88 μg/ml,
cellTracker™ Green CMFDA, Cat.No·C7025, Molecular Probes) was
Fig.1B) was significantly lower than the IC50 value determined for
employed to measure intracellular non-protein thiols [50]. The log
promastigotes (17.80 ± 1.42 μg/ml, Fig. 1A) and was closer to that
phase L. donovani promastigotes (2 × 106 cells/ml) treated with DBA
reported for standard drug miltefosine (0.01–10.9 μg/ml). Chemical
(0–40 μg/ml) for 24 h were collected. The cell pellets were then washed
structures of curcumin (a) and DBA (b) are shown in Fig. 1C. DBA was
with PBS, incubated with CMFDA in the dark for 15 min at 37 °C and
also tested against the BALB/c mouse cell line J774A.1 to determine
analyzed for fluorescence in the FL1 channel, equipped with a 530/
whether the doses used for IC50 determination for intracellular amas-
30 nm band pass filter by using a FACSCalibur flow cytometer and
tigotes were toxic to the cell. This revealed that the 50% cytotoxic dose
analyzed by CellQuestPro software. Ten thousand events from each
(CC50) was higher than the IC50 dose for intracellular amastigotes
sample were acquired to ensure adequate data and at least three in-
(Fig. 1D) with a SI (selectivity index) value of 15.34. To determine the
dependent experiments were carried out.
mechanism of cell death triggered by DBA, we evaluated plasma
membrane integrity in treated promastigotes stained with PI which
2.14. Statistical analysis diffuses across permeable membranes and binds to nucleic acids. This
method has been applied earlier by other investigators [44, 45]. As
Each experiment was performed at least three times and results shown in Fig. 1E, increased cell death along the x-axis (M2) by the
expressed as mean ± SD. The data were analyzed by one-way analysis gradual shifting of the dead cell population (increased MFI) was ob-
of variance using GraphPad Prism program. Multiple comparisons of served with increased concentration of DBA. Treated promastigotes
the means were made through two-way analysis of variance (ANOVA) were stained with PI in a concentration-dependent manner. A sharp
with a Newman–Keuls post-hoc test. Statistically significant difference increase in PI positive (M2) cells from counts 19.12 ± 0 at 10 μg/ml to
between sample and control were designated as: *to denote P < .05 counts 48.70 ± 0 at 20 μg/ml and thereafter gradual decrease from
and **to denote P < .01 levels of confidence while NS denotes dif- counts 44.70 ± 2.84 at 40 μg/ml after 24 h-treatment were observed.
ference that is statistically not significant.

632
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

Fig. 4. [A] DBA causes cell cycle arrest in the G0/G1 phase of L. donovani promastigotes. Cells were treated with different concentrations [a, b, c and d represent 0,
10, 20 and 40 μg/ml respectively] of DBA for 24 h and stained with propidium iodide (PI). The DNA content was analyzed using flow cytometry with Cell Quest
software. Untreated cells were used as control. (e) The bar graph shows mean values and S.D. (*P < .05; **P < .01 compared with the control group, n = 3), NS:
not significant. [B] Externalization of phosphatidylserine in DBA treated promastigotes: L. donovani promastigotes were incubated with different concentrations [a, b,
c and d represent 0, 10, 20 and 40 μg/ml respectively] of DBA for 24 h and analyzed by flow cytometry. After indicated incubation time, a significant number of
membrane compromised cells were stained positively by Annexin V- FITC, PI (upper-right quadrant LA), and only PI positive (upper-left quardrant N). Undamaged
cells were unstained with Annexin V-FITC/PI (bottom left quadrant L). (e) The bar graph shows mean values and S.D. (**P < .01 compared with the control group,
n = 3). [C] DBA-induced DNA fragmentation in promastigotes of L. donovani was analyzed by TUNEL assay using flow cytometry. Increasing concentrations of DBA
showed the increase number of cells (R2) stained by anti-BrdU which have moved from non apoptotic population (G0/G1 and G2 phase) to the apoptotic population
(S phase). Where (a), (b), (c) and (d) show promastigotes treated with different concentrations (0, 10, 20 and 40 μg/ml) of DBA respectively. All data depicted in this
figure are representative of at least three replicates. [D] Measurement of DBA induced changes in intracellular Ca2+ level: Cells were treated with different
concentrations [a, b, c and d represent 0, 10, 20 and 40 μg/ml respectively] of DBA for 24 h and stained with fluo-4 AM. The cytosolic Ca2+ level was analyzed using
flow cytometry with Cell Quest software. Untreated cells were used as control. All data depicted in this figure are representative of at least three replicates.

Untreated cells served as control and showed no cell death. donovani compared with untreated parasites, which showed typical
characteristics with an elongated shape, long flagella and smooth cell
3.2. DBA induced morphological and ultrastructural changes surfaces (Fig. 2a). The Fig. 2b shows alterations in shape and size and
cellular disintegration in the DBA-treated parasites. These cells ex-
For investigation of cell death induced by DBA in promastigotes, hibited shrinking and rounding-up responses and in some cases, mem-
analysis by SEM and TEM was conducted. SEM revealed that DBA brane protrusions resembling surface blebs and ruffling of the plasma
caused morphological alterations in the promastigote forms of L. membrane. This type of morphological effect has previously been

633
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

Fig. 5. [A] Changes in mitochondrial membrane potential following treatment with DBA. L. donovani promastigotes were incubated with different concentrations [a,
b, c and d represent 0, 10, 20 and 40 μg/ml respectively] of DBA for 24 h, loaded in dark for 30 min with Mitotracker (10 μM) and analyzed by flow cutometry. All
data depicted in this figure are representative of at least three replicates. [B] Intracellular level of the glutathione (GSH) in L. donovani promastigotes treated with
different concentrations [a, b, c and d represent 0, 10, 20 and 40 μg/ml respectively] of DBA for 24 h and its fluorescence was measured using a flow cytometer. All
data depicted in this figure are representative of at least three replicates.

shown by other investigators [51]. promastigotes.


In normal promastigotes (Fig. 3 A), all features observed by TEM
were characteristic of Leishmania such as single reticulated mitochon-
3.4. Analysis of externalized phosphatidylserine
dria on the periphery of the cell, an oval nucleus, glycosomes, the rod
shaped kinetoplast disc, and the paraflagellar rod within the flagellar
To investigate the mechanism of DBA-induced apoptotic effects in
pocket [52]. Longitudinal section of the promastigote revealed a deep
Leishmania cells, double staining with Annexin V-FITC and PI was em-
flagellar pocket. The basal body of the flagellum and the closely asso-
ployed. This double staining allows differentiation between early
ciated kinetoplast disc were positioned at the bottom of the flagellar
apoptotic (Annexin V-FITC positive, A), late apoptotic (Annexin V-FITC
pocket as commonly seen in promastigotes. Ultrastructural analysis of
and PI positive, LA), necrotic (PI positive, N) and live cells (unstained)
the DBA-treated promastigotes (Fig. 3B) revealed severe damage to the
[53]. Late exponential-phase promastigotes of L. donovani were treated
parasite mitochondrion. Promastigotes treated with the IC50 con-
with different concentrations (10, 20 and 40 μg/ml) of DBA for 24 h, co-
centration (17.80 ± 3 μg/ml) of DBA for 24 h showed different degrees
stained with PI and Annexin V-FITC, and analyzed by flow cytometry.
of morphological changes which induced loss of parasite viability and
The percentage of DBA treated promastigotes that were positive for
severe loss of cristae and matrix were also seen. Moreover, the matrix
Annexin-V (A + LA) which indicated phosphatidylserine exposure,
displayed a washed-out appearance, indicating a decrease in the elec-
gradually increased, to 7.24 ± 0.24%, 12.66 ± 0.42% and
tron density, Golgi disruption and extensive cytoplasmic vacuolization.
17.36 ± 0.41% at 10, 20and 40 μg/ml, respectively (Fig. 4B). The PI is
an impermeable fluorescent probe that enters cells only when the
permeability of plasma membrane is lost. The number of cells that were
3.3. Cell cycle analysis
PI-positive (upper-left quadrant, N) gradually increased from
8.35 ± 1.68%, 22.92 ± 0.70% and 36.03 ± 0.94% at 10, 20 and
To assess the role of different concentrations (10, 20 and 40 μg/ml)
40 μg/ml, respectively (Fig. 4B). These observations suggested that
of DBA in mediating G0/G1 arrest, cell cycle analysis was performed
these cells were considered to be necrotic. Untreated cells were pri-
based on PI staining using flow cytometry (42). As shown in Fig. 4A,
marily FITC Annexin-V and PI negative, indicating that they were vi-
DBA (at 10, 20 and 40 μg/ml concentrations) induced a marked in-
able and not undergoing apoptosis or necrosis.
crease (~2 fold) in the number of cells in the G0/G1 phase (G1:
43.60 ± 0.98%, 53.65 ± 0.56% and 78.41 ± 0.65%), and simulta-
neous decrease (~3 fold) in both S phase (S: 39.80 ± 1.27%, 3.5. Terminal Deoxynucleotidyltranferase(TdT)-mediated dUTP Nick end
31.84 ± 1.56% and 16.05 ± 0.21%) and G2/M phase labeling (TUNEL) assay
(16.60 ± 2.26%, 14.51 ± 2.13% and 5.54 ± 0.86%) compared with
respective controls (G0/G1: 37.20 ± 1.55%, S: 48.18 ± 2.39% and The TUNEL assay was performed to verify whether DBA was able to
G2/M: 14.62 ± 3.94%). At higher concentration of DBA (40 μg/ml) we induce DNA fragmentation in promastigotes of L. donovani.
found 2.66 ± 0.38% sub-G1 peak which is characteristic of apoptosis Promastigotes when incubated for 24 h with DBA at concentrations of
(Fig. 4Ad). The concentration-dependent effect on G0/G1 arrest in 10, 20 and 40 μg/ml showed small changes in the MFI (1.29 ± 0.03%,
promastigotes was largely at the expense of S phase cells, with lesser 1.91 ± 0.09% and 1.60 ± 0.06% TUNEL-positive cells) when com-
change in the G2/M-phase cell population compared to the untreated pared with untreated promastigotes (0.34 ± 0.04%) (Fig. 4C).

634
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

Increasing the concentrations of DBA showed increased number of cells synthesizable, simple monoketone analog of curcumin without the
(as stained by anti-BrdU) moving from non-apoptotic population (G0/ highly reactive β-diketone moiety and with unsubstituted terminal
G1 and G2 phase) to the apoptotic population (S phase). phenyl rings in its structure to provide stability essential for clinically
desirable pharmacokinetic profile.
3.6. Intracellular Ca2+ level in the DBA-treated promastigotes We for the first time report the antiproliferative effect of this cur-
cumin analog (DBA) in the intracellular amastigotes of L. donovani. The
To assess the variation of cytosolic Ca2+ levels in both treated and observed antiproliferative effects indicated that DBA was more potent
untreated L. donovani promastigotes, Fluo4 was used as a calcium against intracellular amastigotes, the clinically more relevant stage of
fluorescent probe. The data showed a significant increase in fluores- the parasite than its promastigotes stage. On comparison with reference
cence attributable to the cytosolic Ca2+ ion content in promastigotes drug miltefosine, which is in clinical use in the endemic areas of India,
with increasing concentrations of DBA (MFI: 45.52 ± 1.48 at 20 μg/ml DBA showed IC50 value (7.43 μg/ml) closer to that reported for milte-
and 67.86 ± 1.79 at 40 μg/ml) as compared to control (MFI: fosine (0.01–10.9 μg/ml)(https://www.accessdata.fda.gov/drugsatfda_
30.07 ± 0.15) (Fig. 4D). However, the low concentration of DBA docs/nda/2014/204684Orig1s000MicroR.pdf). DBA showed no cyto-
(10 μg/ml) failed to change the Ca2+ level in promastigotes (MFI: toxicity against the BALB/c mouse cell line J734A.1.
32.50 ± 1.78). Alterations in morphology of the DBA-treated parasites were as-
sessed by SEM which showed remarkable changes when compared to
3.7. Measurement of mitochondrial membrane potential untreated cells. The main ultrastructural alteration was observed in the
mitochondrion-kinetoplast complex, which showed intense mitochon-
TEM study revealed that DBA induced significant changes and da- drial swelling with an increase in the number of cristae. DBA changed
mage in promastigote mitochondria. Based on this, membrane potential the profile of the cell cycle with an increase in G0/G1 population as-
of treated promastigotes was determined by flow cytometry using a sociated with a decrease in S (synthesis) and G2/M phases when
cell-permeable dye, MitoTracker deep red. Treatment with different compared to control. Although it has been established by Weingartner
concentrations (0–40 μg/ml) of DBA induced decrease in the MFI et al. [60] that Leishmania promastigotes lack phosphatidylserine, An-
(158.36 ± 1.60, 139.02 ± 0.07 and 137.14 ± 0.13 respectively) re- nexin-V binding as an early indicator of apoptosis has been used by
lative to the untreated promastigotes (224.97 ± 1.44) (Fig. 5A). Other other investigators [61]. As shown in Fig. 4, L. donovani parasites upon
investigator has shown that mitochondrial membrane dissipation may treatment with DBA do not indicate early apoptotic characteristics. If
be caused due to increase in cytosolic Ca2+ level [54]. Since depolar- they had shown early apoptotic features, then we would have obtained
ization of mitochondrial membrane potential is an important event in Annexin+/PI−. Instead, we obtained PI+ and then Annexin+\PI+,
apoptosis, we investigated the increase in cytosolic Ca2+ level, which is which means that the parasites enter into late apoptotic stage without
upstream event of mitochondrial membrane dissipation, to confirm going into early apoptotic phase and these cells have lost their mem-
apoptosis. brane integrity. DBA caused an increase in cytosolic Ca+2 level leading
to mitochondrial membrane potential dissipation [54]. We further
3.8. Measurement of intracellular non-protein thiols correlated loss of membrane potential with depletion of antioxidant
molecules such as intracellular non-protein thiol (GSH).
Reduced glutathione (GSH) is an important cellular component for Our results demonstrate that DBA has potent anti-prolific activity
protecting kinetoplastids, such as Leishmania from ROS and it is a against L. donovani. It alters the general ultrastructure and the mi-
component of trypanothione, a major antioxidant of this parasite [55, tochondrial physiology of the parasite and triggers apoptotic cell death.
56]. Accordingly, the effect of DBA on the level of this reduced in- In conclusion, based on its potent in vitro antiparasitic activity against L.
tracellular non-protein thiol was assessed using fluorescent probe, donovani, this study has identified DBA, a simple curcumin analog as a
CMFDA. Promastigotes treated with DBA at different concentrations potential candidate for further investigations on leishmaniasis. With its
(0–40 μg/ml) showed remarkable decrease in the level of GSH from low toxicity profile, DBA compares favorably, at least in vitro, with
untreated (MFI: 278.54 ± 1.02) to treated cells (MFI: 60.52 ± 0.40, existing drug miltefosine and is a suitable candidate for further in vivo
48.82 ± 0.12 and 52.45 ± 0.31) (Fig. 5B). investigations against VL.
From the above-mentioned results, we can conclude that DBA
caused depolarization of the mitochondrion and the depletion of re- Conflict of interest
duced glutathione, triggering an apoptotic response in L. donovani
promastigotes. The authors declare no conflict of interest.

4. Discussion Acknowledgments

Cytotoxic and antiparasitic effects of curcumin on protozoan para- The authors are grateful to Mr. A.L. Vishwakarma for assistance
sites have been demonstrated earlier in cultures against Trypanosoma, with flow cytometry. This work was supported by Council of Scientific
Giardia, and Plasmodium falciparum [57–59]. It has been reported to be and Industrial Research (CSIR) (BSC0114) funded network project
cytotoxic to several strains of Leishmania including L. major, L. tropica L. “Host Interactome analysis: Understanding the Role of Host molecules
infantum [29] and L. donovani [21]. Limited investigations have been in Parasitic Infection (HOPE).” This is CDRI communication no. 196/
carried out to decipher the mode of cell death using curcumin as a 2015/NS. While this manuscript was under preparation, Prof. Dr.
leishmaniacidal agent [21]. There is only one reported study estab- Govind J. Kapadia passed away on August 23, 2016. He will be re-
lishing apoptosis as the main mechanism of cell death in Leishmania membered by students, faculty and co-workers as an accomplished
parasites in response to curcumin [13]. scientist, dedicated teacher, inspiring mentor and loyal friend.
Among the curcumin structural modifications evaluated, elimina-
tion of the hydrolysis-prone diketo functionality and incorporating a References
range of alternate substituents on the terminal phenyl rings have re-
sulted in new synthetic curcuminoids with improved structural stability [1] A.H. Lopes, T. Souto-Padrón, F.A. Dias, M.T. Gomes, G.C. Rodrigues,
L.T. Zimmermann, T. Alves e Silva, A.B. Vermelho, Trypanosomatids: odd organ-
and increased tissue uptake under both in vitro and in vivo conditions
isms, devastating diseases, Open Parasitol. J. 4 (2010) 30–59.
resulting in enhanced bioavailability [24, 25]. In the present study, we [2] P.D. Ready, Epidemiology of visceral leishmaniasis, Clin. Epidemiol. 6 (2014) 147.
have investigated the leishmanicidal activity of DBA, an easily [3] W.H.O, Control of the leishmaniases, World Health Organization Technical Report

635
I.S. Chauhan et al. Parasitology International 67 (2018) 627–636

Series, 949 2010, p. xii. experimental model, Lab. Investig. 88 (12) (2008) 1329–1339.
[4] A.R. Zink, M. Spigelman, B. Schraut, C.L. Greenblatt, A.G. Nerlich, H.D. Donoghue, [35] Y. Chen, E. Mcmillan-Ward, J. Kong, S.J. Israels, S.B. Gibson, Mitochondrial elec-
Leishmaniasis in ancient Egypt and upper Nubia, Emerg. Infect. Dis. 12 (10) (2006) tron-transport-chain inhibitors of complexes I and II induce autophagic cell death
1616. mediated by reactive oxygen species, J. Cell Sci. 120 (23) (2007) 4155–4166.
[5] B.L. Herwaldt, Leishmaniasis, Lancet 354 (9185) (1999) 1191–1199. [36] T. Kobets, I. Grekov, M. Lipoldova, Leishmaniasis: prevention, parasite detection
[6] W.H.O, Report of the Fifth Consultative Meeting on Leishmania/HIV Coinfection, and treatment, Curr. Med. Chem. 19 (10) (2012) 1443–1474.
Addis Ababa, Ethiopia, (2007), pp. 20–22. [37] L.A. Hull, The Dibenzalacetone reaction revisited, J. Chem. Educ. 78 (2) (2001)
[7] L.H. Freitas-Junior, E. Chatelain, H.A. Kim, J.L. Siqueira-Neto, Visceral leishma- 226.
niasis treatment: what do we have, what do we need and how to deliver it? Int. J. [38] B.L. Hawbecker, D.W. Kurtz, T.D. Putnam, P.A. Ahlers, G.D. Gerber, Aldol con-
Parasitol. 2 (2012) 11–19. densation: a simple teaching model for organic laboratory, J. Chem. Educ. 55 (8)
[8] M. Bessat, S. El Shanat, Leishmaniasis: epidemiology, control and future perspec- (1978) 540.
tives with special emphasis on Egypt, J. Trop. Dis. Public Health 2 (2) (2015). [39] S.L.F. Jones, Synthesis of Bioactive Distyrylketones and Isolation of Acetogenins
[9] A. Clem, A current perspective on leishmaniasis, J. Global Infect. Dis. 2 (2) (2010) from Annona senegalensis, Pharmaceutical Chemistry, Howard University,
124. Washington, DC, USA, 1997 (Doctoral thesis).
[10] S.L. Croft, K. Seifert, V. Yardley, Current scenario of drug development for leish- [40] A.F. Cockerill, D.M. Rackham, Kinetics of reduction of 1, 5-diarylpenta-1, 4-dien-3-
maniasis, Indian J. Med. Res. 123 (3) (2006) 399. ones with sodium borohydride in propan-2-ol, J. Chem. Soc. Perkin Trans. 2 (14)
[11] J. Chakravarty, S. Sundar, Drug resistance in leishmaniasis, J. Global Infect. Dis. 2 (1972) 2076–2081.
(2) (2010) 167. [41] A.A. Zahir, I.S. Chauhan, A. Bagavan, C. Kamaraj, G. Elango, J. Shankar, N. Arjaria,
[12] P.L. Olliaro, Drug combinations for visceral leishmaniasis, Curr. Opin. Infect. Dis. S.M. Roopan, A.A. Rahuman, N. Singh, Green synthesis of silver and titanium di-
23 (6) (2010) 595–602. oxide nanoparticles using Euphorbia prostrata extract shows shift from apoptosis to
[13] B.B. Aggarwal, C. Sundaram, N. Malani, H. Ichikawa, Curcumin: the indian solid G0/G1 arrest followed by necrotic cell death in Leishmania donovani, Antimicrob.
gold, in: B.B. Aggarwal, Y.J. Surh, S. Shishodia (Eds.), The Molecular Targets and Agents Chemother. 59 (8) (2015) 4782–4799.
Therapeutic Uses of Curcumin in Health and Disease, Advances in Experimental [42] G. Chouhan, M. Islamuddin, F. Ahmad, D. Sahal, F. Afrin, Antileishmanial potential
Medicine and Biology, Springer, Boston, MA, 2007, pp. 1–75. of Piper nigrum seed extracts against Leishmania donovani, Open J. Med.
[14] S. Zorofchian Moghadamtousi, H. Abdul Kadir, P. Hassandarvish, H. Tajik, Microbiol. 4 (04) (2014) 228.
S. Abubakar, K. Zandi, A review on antibacterial, antiviral, and antifungal activity [43] T. Riss, R. Moravec, Simplifying cytotoxicity screening, Promega Notes 83 (2003)
of curcumin, BioMed Res. Int. 53 (2014) 186864, , http://dx.doi.org/10.1155/ 10–13.
2014/186864 (1–12). [44] M.K. Gould, X.L. Vu, T. Seebeck, H.P. de Koning, Propidium iodide-based methods
[15] R. De, P. Kundu, S. Swarnakar, T. Ramamurthy, A. Chowdhury, G.B. Nair, for monitoring drug action in the kinetoplastidae: comparison with the Alamar blue
A.K. Mukhopadhyay, Antimicrobial activity of curcumin against Helicobacter pylori assay, Anal. Biochem. 382 (2) (2008) 87–93.
isolates from India and during infections in mice, Antimicrob. Agents Chemother. [45] U. Gaur, M. Showalter, S. Hickerson, R. Dalvi, S.J. Turco, M.E. Wilson,
53 (4) (2009) 1592–1597. S.M. Beverley, Leishmania donovani lacking the Golgi GDP-Man transporter LPG2
[16] H. Hatcher, R. Planalp, J. Cho, F. Torti, S. Torti, Curcumin: from ancient medicine exhibit attenuated virulence in mammalian hosts, Exp. Parasitol. 122 (3) (2009)
to current clinical trials, Cell. Mol. Life Sci. 65 (11) (2008) 1631–1652. 182–191.
[17] A. Shehzad, F. Wahid, Y.S. Lee, Curcumin in cancer chemoprevention: molecular [46] J. Kaur, B.K. Singh, R.P. Tripathi, P. Singh, N. Singh, Leishmania donovani: a gly-
targets, pharmacokinetics, bioavailability, and clinical trials, Arch. Pharm. 343 (9) cosyl dihydropyridine analogue induces apoptosis like cell death via targeting
(2010) 489–499. pteridine reductase 1 in promastigotes, Exp. Parasitol. 123 (3) (2009) 258–264.
[18] N. Chainani-Wu, Safety and anti-inflammatory activity of curcumin: a component of [47] J. Rodrigues, R.D. Silva, H. Noronha, A. Pedras, H. Gerós, M. Côrte-Real, Flow
tumeric (Curcuma longa), J. Alter. Complement. Med. 9 (1) (2003) 161–168. cytometry as a novel tool for structural and functional characterization of isolated
[19] P. Anand, S.G. Thomas, A.B. Kunnumakkara, C. Sundaram, K.B. Harikumar, yeast vacuoles, Microbiology 159 (Pt 5) (2013) 848–856.
B. Sung, S.T. Tharakan, K. Misra, I.K. Priyadarsini, K.N. Rajasekharan, Biological [48] R. Sen, P. Saha, A. Sarkar, S. Ganguly, M. Chatterjee, Iron enhances generation of
activities of curcumin and its analogues (congeners) made by man and mother free radicals by Artemisinin causing a caspase-independent, apoptotic death in
nature, Biochem. Pharmacol. 76 (11) (2008) 1590–1611. Leishmania donovani promastigotes, Free Radic. Res. 44 (11) (2010) 1289–1295.
[20] Y.-J. Shang, X.-L. Jin, X.-L. Shang, J.-J. Tang, G.-Y. Liu, F. Dai, Y.-P. Qian, G.-J. Fan, [49] R.P. Haugland, M.T. Spence, I.D. Johnson, Handbook of Fluorescent Probes and
Q. Liu, B. Zhou, Antioxidant capacity of curcumin-directed analogues: structure–- Research Chemicals, Molecular Probes, Eugene, OR, USA, 1996.
activity relationship and influence of microenvironment, Food Chem. 119 (4) [50] A. Sarkar, G. Mandal, N. Singh, S. Sundar, M. Chatterjee, Flow cytometric de-
(2010) 1435–1442. termination of intracellular non-protein thiols in Leishmania promastigotes using 5-
[21] R. Das, A. Roy, N. Dutta, H.K. Majumder, Reactive oxygen species and imbalance of chloromethyl fluorescein diacetate, Exp. Parasitol. 122 (4) (2009) 299–305.
calcium homeostasis contributes to curcumin induced programmed cell death in [51] F.A. Marinho, K. Gonçalves, S.S. Oliveira, D.S. Gonçalves, F.P. Matteoli,
Leishmania donovani, Apoptosis 13 (7) (2008) 867–882. S.H. Seabra, A.C.S. Oliveira, M. Bellio, S.S. Oliveira, T. Souto-Padrón, The calpain
[22] L. Cui, J. Miao, L. Cui, Cytotoxic effect of curcumin on malaria parasite Plasmodium inhibitor MDL28170 induces the expression of apoptotic markers in Leishmania
falciparum: inhibition of histone acetylation and generation of reactive oxygen amazonensis promastigotes, PLoS One 9 (1) (2014).
species, Antimicrob. Agents Chemother. 51 (2) (2007) 488–494. [52] W. De Souza, From the cell biology to the development of new chemotherapeutic
[23] Y. Sugiyama, S. Kawakishi, T. Osawa, Involvement of the β-diketone moiety in the approaches against trypanosomatids: dreams and reality, Kinet. Biol. Dis. 1 (1)
antioxidative mechanism of tetrahydrocurcumin, Biochem. Pharmacol. 52 (4) (2002) 3.
(1996) 519–525. [53] A. Efstathiou, N. Gaboriaud-Kolar, D. Smirlis, V. Myrianthopoulos,
[24] A.A. Alkhaldi, D.J. Creek, H. Ibrahim, D.-H. Kim, N.B. Quashie, K.E. Burgess, K. Vougogiannopoulou, A. Alexandratos, M. Kritsanida, E. Mikros, K. Soteriadou,
C. Changtam, M.P. Barrett, A. Suksamrarn, H.P. de Koning, Potent trypanocidal A.-L. Skaltsounis, An inhibitor-driven study for enhancing the selectivity of in-
curcumin analogs bearing a monoenone linker motif act on Trypanosoma brucei by dirubin derivatives towards leishmanial Glycogen Synthase Kinase-3 over leish-
forming an adduct with trypanothione, Mol. Pharmacol. 87 (3) (2015) 451–464. manial cdc2-related protein kinase 3, Parasit. Vectors 7 (1) (2014) 234.
[25] D. Shetty, Y.J. Kim, H. Shim, J.P. Snyder, Eliminating the heart from the curcumin [54] S.B. Mukherjee, M. Das, G. Sudhandiran, C. Shaha, Increase in cytosolic Ca2+ levels
molecule: monocarbonyl curcumin mimics (MACs), Molecules 20 (1) (2014) through the activation of non-selective cation channels induced by oxidative stress
249–292. causes mitochondrial depolarization leading to apoptosis-like death in Leishmania
[26] G. Liang, L. Shao, Y. Wang, C. Zhao, Y. Chu, J. Xiao, Y. Zhao, X. Li, S. Yang, donovani promastigotes, J. Biol. Chem. 277 (27) (2002) 24717–24727.
Exploration and synthesis of curcumin analogues with improved structural stability [55] G. Sudhandiran, C. Shaha, Antimonial-induced increase in intracellular Ca2+
both in vitro and in vivo as cytotoxic agents, Bioorg. Med. Chem. 17 (6) (2009) through non-selective cation channels in the host and the parasite is responsible for
2623–2631. apoptosis of intracellular Leishmania donovani amastigotes, J. Biol. Chem. 278 (27)
[27] S. Handayani, I.S. Arty, Synthesis of hydroxyl radical scavengers from benzalace- (2003) 25120–25132.
tone and its derivatives, J. Phys. Sci. 19 (2) (2008) 61–68. [56] F. Fonseca-Silva, J. Inacio, M.M. Canto-Cavalheiro, E.E. Almeida-Amaral, Reactive
[28] M. Shahiduzzaman, A. Daugschies, Curcumin: a natural herb extract with anti- oxygen species production and mitochondrial dysfunction contribute to quercetin
parasitic properties, in: H. Mehlhorn (Ed.), Nature Helps: How Plants and Other induced death in Leishmania amazonensis, PLoS One 6 (2) (2011) e14666.
Organisms Contribute to Solve Health Problems. Parasitology Research [57] M. Nose, T. Koide, Y. Ogihara, Y. Yabu, N. Ohta, Trypanocidal effects of curcumin in
Monographs 1, Springer, Berlin, 2011, pp. 141–151. vitro, Biol. Pharm. Bull. 21 (6) (1998) 643–645.
[29] D. Saleheen, S.A. Ali, K. Ashfaq, A.A. Siddiqui, A. Agha, M.M. Yasinzai, Latent ac- [58] L. Perez-Arriaga, M. Mendoza-Magana, R. Cortes-Zarate, A. Corona-Rivera,
tivity of curcumin against leishmaniasis in vitro, Biol. Pharm. Bull. 25 (3) (2002) L. Bobadilla-Morales, R. Troyo-Sanromán, M. Ramirez-Herrera, Cytotoxic effect of
386–389. curcumin on Giardia lamblia trophozoites, Acta Trop. 98 (2) (2006) 152–161.
[30] M.M.-Y. Chan, N.S. Adapala, D. Fong, Curcumin overcomes the inhibitory effect of [59] H.B. Rasmussen, S.B. Christensen, L.P. Kvist, A. Karazmi, A simple and efficient
nitric oxide on Leishmania, Parasitol. Res. 96 (1) (2005) 49–56. separation of the curcumins, the antiprotozoal constituents of Curcuma longa,
[31] C. Changtam, H.P. de Koning, H. Ibrahim, M.S. Sajid, M.K. Gould, A. Suksamrarn, Planta Med. 66 (4) (2000) 396–398.
Curcuminoid analogs with potent activity against Trypanosoma and Leishmania [60] A. Weingärtner, G. Kemmer, F.D. Müller, R.A. Zampieri, M.G. dos Santos,
species, Eur. J. Med. Chem. 45 (3) (2010) 941–956. J. Schiller, T.G. Pomorski, Leishmania promastigotes lack phosphatidylserine but
[32] D.C. Gomes, L.V. Alegrio, M. De Lima, L.L. Leon, C. Araujo, Synthetic derivatives of bind annexin V upon permeabilization or miltefosine treatment, PLoS One 7 (8)
curcumin and their activity against Leishmania amazonensis, (2012) e42070.
Arzneimittelforschung 52 (2) (2001) 120–124. [61] S. Chowdhury, T. Mukherjee, S.R. Chowdhury, S. Sengupta, S. Mukhopadhyay,
[33] R.F. Tayyem, D.D. Heath, W.K. Al-Delaimy, C.L. Rock, Curcumin content of tur- P. Jaisankar, H.K. Majumder, Disuccinyl betulin triggers metacaspase-dependent
meric and curry powders, Nutr. Cancer 55 (2) (2006) 126–131. endonuclease G-mediated cell death in unicellular protozoan parasite Leishmania
[34] N. Adapala, M.M. Chan, Long-term use of an antiinflammatory, curcumin, sup- donovani, Antimicrob. Agents Chemother. 58 (4) (2014) 2186–2201.
pressed type 1 immunity and exacerbated visceral leishmaniasis in a chronic

636

You might also like