You are on page 1of 12

Biomedicine & Pharmacotherapy 111 (2019) 224–235

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Serine protease inhibitors rich Coccinia grandis (L.) Voigt leaf extract induces T
protective immune responses in murine visceral leishmaniasis

Asmita Pramanik, Dibyendu Paik, Pijush Kanti Pramanik, Tapati Chakraborti
Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, 741235, West Bengal, India

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

Keywords: Leishmaniasis is a parasite-mediated tropical disease affecting millions of individuals worldwide. The available
Antileishmanial activity antileishmanial chemotherapeutic modalities exhibit adverse toxicity, exorbitant price and advent of drug-re-
Coccinia grandis (L.) Voigt sistant parasites. Hence, plant-derived products are an alternative preference for the emergence of novel and
Immunomodulation effective antileishmanial agents that rejuvenate the host immunity with limited toxicity. The present work is
Leishmania donovani
complementary to our previous report that revealed the in vitro antileishmanial and immunomodulatory activity
Serine protease inhibitor
Visceral leishmaniasis
of Coccinia grandis (L.) Voigt leaf extract (Cg-Ex) rich in serine protease inhibitors. Thus, preliminary objectives
of the study were to elucidate the leishmanicidal activity and host effector mechanism in Leishmania donovani
infected BALB/c mice treated with Cg-Ex. Oral administration of Cg-Ex significantly reduced the spleen and liver
parasite burden at dose-dependently. The parasite elimination was associated with generation of ROS and NO
that are interrelated with up-regulation of disease-suppressing Th1 cytokines and down-regulation of disease-
promoting Th2 cytokines at both protein and mRNA level. Moreover, Cg-Ex augmented the delayed-type hy-
persensitivity (DTH) response and serum IgG2a level which are correlated with the diminution of parasite
burden with no hepatic and renal toxicity. Additionally, histological analysis of spleen depicted the improvement
of structural disorganization of white and red pulp after Cg-Ex treatment. Therefore, our intriguing findings have
presented the first indication of in vivo antileishmanial efficacy through activation of pro-inflammatory immune
responses of the host by a natural plant leaf extract (Cg-Ex) containing serine protease inhibitors which could
have a role as a potential immunomodulator against visceral leishmaniasis.

1. Introduction leishmaniasis (PKDL) remain as major problems to eliminate the VL


from India [2]. The emergence of novel and promising antileishmanial
Leishmania parasite belongs to the order of Kinetoplastid and resides medicine has a paramount preference on the ground that prevention of
in the phagolysosome of phagocytic cells by subverting the mammalian the disease is very challenging. It is well studied that Leishmania spp.
host immune system. The parasite develops a wide array of clinical deactivate the defence machinery of the host macrophage cells and
manifestations ranging from the asymptomatic cutaneous leishmaniasis propagate disease-associated symptoms. Thus, development of the new
(CL) to the fatal visceral leishmaniasis (VL), which affects around 350 chemotherapeutic approach against VL especially focuses on the direct
million individuals in 98 endemic countries and territories [1]. Until leishmanicidal effect and concurrent stimulation of the host’s protective
now, treatment strategies of VL have been primarily devoted to che- immune system with minimal toxicity [3].
motherapy due to the deficiency of commercially available effective Proteases play an essential role in the various biological processes of
vaccines. However, the limited option of antileishmanial drugs, devel- human as well as protozoan parasites. In Leishmania parasites, proteases
opment of multiple drug-resistant parasite strains, relapse of the disease exhibit a putative function for their proliferation, life cycle, and also act
even with HIV-VL co-infection and the onset of post-kala-azar dermal as a very important virulence factor in the propagation of infection.

Abbreviations: AST, aspartate transaminase; ALT, alanine transaminase; Cg-Ex, Coccinia grandis (L.) Voigt leaf extract; CL, cutaneous leishmaniasis; DTH, delayed-
type hypersensitivity; FCS, fetal calf serum; IFN-γ, interferon-gamma; IgG1, immunoglobulin G1; IgG2a, immunoglobulin G2a; IL-10, interleukine-10; IL-12, in-
terleukine-12; iNOS, inducible nitric oxide synthase; LD-SP, Leishmania donovani serine protease; NO, nitric oxide; PBS, phosphate buffer saline; RNS, reactive
nitrogen species; ROS, reactive oxygen species; SAG, sodium stibogluconate; SLA, soluble leishmanial antigen; TGF-β, transforming growth factor-beta; TNF-α,
tumour necrosis factor-alpha; VL, visceral leishmaniasis

Corresponding author.
E-mail address: tcbiochem@gmail.com (T. Chakraborti).

https://doi.org/10.1016/j.biopha.2018.12.053
Received 4 September 2018; Received in revised form 10 December 2018; Accepted 14 December 2018
0753-3322/ © 2018 Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Additionally, Leishmania proteases (metallo, cysteine and serine) par- The virulence of promastigotes was maintained in BALB/c mice. They
ticipate in the establishment of the infection by modulating various were infected with the late stationary phase of second passage pro-
processes of the host systems among them the deactivation of host mastigotes, and virulent promastigotes were obtained from infected
immune system is one of the most important events. So, the use of spleen after transforming amastigotes. Subculture of promastigotes
protease inhibitors targeting these proteases which are automatically were passed after 72 h at 22 °C in medium 199 (M199, Gibco) with
affects the protease mediated immune mechanism and block the pro- Hank’s salt containing HEPES (Sigma-Aldrich), 100 U/ml penicillin
pagation of infection [4,5]. Our previous findings of a virulence factor (Gibco), and 100 mg/ml streptomycin (Gibco) supplemented with 10%
from the Indian strain of Leishmania donovani is an intracellular serine (v/v) heat-inactivated fetal calf serum (FCS, Gibco) [16].
protease (58 kDa) which makes provision of an additional objective for
drug targeting aspect [6–8]. Thus, investigation of protease inhibitor
from natural sources could be beneficial to develop a unique protease- 2.3. Ethical clearance
based treatment strategy for combating visceral leishmaniasis.
Plant, is a precious wealth of nature, have a gigantic reservoir of To perform various experiments on the BALB/c mouse model, the
novel bioactive materials and these are alternative and attractive op- ethical clearance was obtained from Institutional Animal Ethics
tions for the drug discovery. Hence, plant-derived products can be Committee (IAEC), Department of Zoology (Registration number:892/
considered as a glimmer of hope for advancement of the chemother- GO/Re/S/01/CPCSBA, Pr. 20.04.2014-28.04.2019), University of
apeutic approach in different diseases. Currently, many medicinal plant Kalyani, Kalyani, West Bengal, India.
derived natural-products are used as the drug candidates for various
diseases [9]. Several scientific studies in recent years have suggested
the efficacy of different plant extracts and purified components against 2.4. Animals
fungus, protozoan parasites, helminths and other diseases [10]. As
noted that plant kingdom represents an excellent source of endogenous Adult female BALB/c mice 4–6 weeks (20–25 g body weight) were
protease inhibitors along with numerous secondary metabolites and selected as in vivo model and maintained in the animal house of
plant-based protease inhibitors are recently found to be efficient for Department of Zoology, University of Kalyani, Kalyani, West Bengal,
cancer, cardiovascular, and inflammatory diseases. In addition, plant India, according to the animal ethics committee guidelines. They were
serine protease inhibitors particularly isolated from leaves and seeds kept at a standard temperature 20 ± 5 °C on a 12 h day and night cycle
have also been studied in the search of alternative therapeutic agents of and were provided standard food pallet diet and water ad libitam.
these diseases [11]. BALB/c mice were used for the isolation of splenocytes and main-
Coccinia grandis (L.) Voigt belongs to the Cucurbitaceae plant family, tenance of the virulent parasites as well as for the determination of in
a climbing perennial tropical medicinal plant [12]. In the Indian tra- vivo antileishmanial efficacy of Cg-Ex [16].
ditional medicine, leaves of this plant have been used to treat diabetes
and also prevent gonorrhoea over a century. Furthermore, serine pro-
tease inhibitor from C. grandis leaf shows the evidence of antibacterial 2.5. In vivo infection and treatment regiment
and antifungal activity [13]. In our earlier reports, serine protease in-
hibitors enriched C. grandis leaf extract (Cg-Ex) exhibited anti- Each female BALB/c mouse was infected through intracardiac route
proteolytic activity against Leishmania serine protease (SP-Ld) and has with 1 × 107 virulent stationary-phase L. donovani promastigotes [17].
also shown in vitro antileishmanial activity against L. donovani with a The experimental sets were divided into eight groups (n = 3 mice/
negligible cytotoxic effect on macrophages [14,15]. Thus, consequences group) such as uninfected control, normal mice with Cg-Ex, infected
of in vivo study with Cg-Ex might acquire significant importance for its control, vehicle control, positive control, Group I, Group II, Group III.
identity as a potent antileishmanial agent. Our primary objective of After 30days of post-infection, infected animals were assigned into
present study was to exploit the antileishmanial efficacy and im- three categories Group I (12 mg/kg body weight/day), Group II (24 mg/
munomodulatory role of Cg-Ex on L. donovani infected BALB/c mice for kg body weight/day), Group III (48 mg/kg body weight/day) and re-
the anticipation of developing an innovative antileishmanial agent for ceived Cg-Ex by oral gavage for consecutive 15days. Another group of
visceral leishmaniasis. normal mice were orally received 48 mg/kg body weight/day of Cg-Ex
for consecutive 15days. Efficacy of Cg-Ex will be evaluated at 1day and
2. Materials & methods 15days of post-treatment by measuring different parameters. Vehicle
control mice were orally received sterile PBS during the treatment
2.1. Preparation of Coccinia grandis (L.) Voigt leaf extract (Cg-Ex) schedule. Sodium stibogluconate (SAG, 20 mg/kg body weight/day for
15days) was used as a positive control (Table 1).
Fresh leaves from Coccinia grandis (L.) Voigt were collected from
University garden and extracted by physical method in 100 mM phos- Table 1
phate buffer, pH = 7.6 (1:5 w/v) at 4 °C, followed by centrifugation at Description of groups used in the in vivo experiment.
8000×g for 30 min and supernatant (crude extract) was collected. Then
Serial No. Group Name Dosage
crude extract was fractionated by ammonium sulfate precipitation 1 Uninfected Control Age-match control
(20–85% saturation) and the precipitate was recovered by centrifuga- 2 Normal control+†Cg-Ex 48mg/kg body weight †Cg-Ex for 15days oral
tion at 10,000×g for 20 min at 4 °C. The precipitate was resuspended in administration in normal mice
10 mM phosphate buffer, pH = 7.6. Then dialysed against 10 mM 3 Infected Control 1×107 L. donovani promastigotes by cardiac puncture
4 Vehicle Control Sterile #PBS treated
phosphate buffer saline (PBS), pH = 7.6 for 48 h at 4 °C and after that
5 Positive Control 20mg/kg body weight §SAG for 15days intravenous administration
lyophilized. Subsequently, lyophilized sample was dissolved in 10 mM 6 Group I 12mg/kg body weight †Cg-Ex for 15days oral administration in infected
phosphate buffer saline, pH = 7.6 and was stored at −20 °C until used mice
[14]. 7 Group II 24mg/kg body weight †Cg-Ex for 15days oral administration in infected
mice
8 Group III 48mg/kg body weight †Cg-Ex for 15days oral administration in
2.2. Parasite cultures infected mice

Indian strain of pentavalent antimony-responsive Leishmania dono- †


Cg-Ex=Coccinia grandis (L.) Voigt leaf extract, PBS = Phosphate buffer saline,
vani promastigotes (MHOM/IN/1983/AG83) was used for this work. SAG = Sodium stibogluconate.

225
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

2.6. Soluble leishmanial antigen (SLA) preparation Corporation, Forma II series). Then, splenocytes were incubated with
5μM DAF-2DA (Sigma-Aldrich) for 20 min at 37 °C in dark. The flow
For the measurement of nitrite, superoxide and cytokines, soluble cytometer (FACS Calibur, Becton Dickinson, USA) was used for mea-
leishmanial antigen (SLA) was prepared from stationary phase L. do- surement of NO and fluorescence intensity was quantified by using FL-1
novani promastigotes according to the standard protocol [18]. Briefly, green filter. Here, data analysis was done by CellQuestPro software
promastigotes (1 × 109/ml) were washed twice with sterile PBS. Then [21].
the lysate was prepared by six cycles of freezing (−70 °C) and thawing
(37 °C) followed by the 5 min incubation on the ice. The partially lysed 2.11. Quantification of Th1/Th2 cytokines by ELISA
promastigotes were fully disrupted by a sonicator thrice for 30 s each
and centrifuged at 10,000 rpm for 30 min at 4 °C. The supernatant Production of Th1 and Th2 cytokines from splenocytes of the dif-
containing soluble antigen was collected and the protein concentration ferent sets of BALB/c mice were quantitatively measured by sandwich
was determined by BCA assay method (Pierce, Thermo Fisher Scien- ELISA. Briefly, isolated splenocytes (1 × 106 /ml) from all mice were
tific). SLA was stored at −20 °C until further use. seeded in 96-well plates and stimulated with SLA (50 μg/ml) for 72 h at
37 °C CO2 incubator (Thermo Electron Corporation, Forma II series).
2.7. Serum collection The released pro-inflammatory and anti-inflammatory cytokines from
the splenocytes were measured in the supernatant as per manufacturer’s
Blood was collected by cardiac puncture after anaesthetization of all instruction using commercial ELISA kits (BD Bioscience, USA). The
groups of BALB/c mice. Then serum was collected by centrifugation at detection limit of these assays was 2.0, 2.5, 5.1, 1.5 and 4.6 pg/ml for
1000 rpm for 5 min at room temperature and stored at −80 °C to per- IL-12, TNF-α, IFN-γ, IL-10 and TGF-β respectively [17].
form the quantitative analysis of IgG1, IgG2a and various biochemical
markers [19]. 2.12. RNA isolation and semi-quantitative RT-PCR analysis of cytokine
genes
2.8. Enumeration of in vivo parasite burden
Total RNA from the isolated splenocytes of different groups of
BALB/c mice from different groups were sacrificed for enumeration BALB/c mice was extracted according to the standard Ribozol method
of parasites in liver and spleen at 1day and 15days of post-treatment. (Life Science Amresco). The first strand cDNA synthesis was done as per
Spleen and liver of each animal from respective groups were excised the protocol provided by qScript cDNA synthesis kit (Quanta
and weighted. Thereafter, spleen and liver parasite burden was mi- Biosciences) and aliquots of 1 μg total RNA were used in 20 μl reaction
croscopically determined from Giemsa (Sigma-Aldrich) stained spleen volume. Then PCR amplification was performed in a Thermal Cycler
and liver imprints respectively and expressed as Leishman Donovan (Bio-Rad, USA) using iProof High-Fidelity Master Mix (Bio-Rad, USA) of
Units (LDU) of Stauber by the following formula [20]: 50 μl reaction mixture. PCR products were subjected on 1.5% agarose
gels and visualized under UV illumination after staining with ethidium
LDU = (No. of amastigotes/1000 cell nuclei) × Weight of organ (in
bromide (Bio-Rad Gel-Doc, USA). The forward and reverse primer se-
mg)
quence of different genes are given in Table 2.

2.9. Measurement of intracellular reactive oxygen species (ROS) production 2.13. Assessment of delayed-type hypersensitivity (DTH) response

Intracellular ROS generation induced by Cg-Ex was measured using Delayed-type hypersensitivity (DTH) response is a salient feature of
cell-permeable probe H2DCFDA in different groups of mice. Splenocytes cell-mediated immunity (CMI) in animal model. According to Kaur et al
were isolated from the individual animal of all experimental groups as DTH response was measured in every group of mice [19]. In brief, all
per the standard protocol [17] and plated in phenol red-free RPMI the groups of mice were subcutaneously injected with SLA in the right
medium and incubated with (50 μg/ml) SLA for 72 h at 37 °C CO2 in- footpad and PBS in their left footpads, 48 h prior to the euthanisation.
cubator (Thermo Electron Corporation, Forma II series). After that, Before sacrificing mice, the thickness of both footpads was measured
splenocytes were incubated with 10μM H2DCFDA (Sigma-Aldrich) for with a Vernier calliper. Results were expressed in diameter of the swell
20 min at 37 °C in dark and production of ROS was measured in terms of region of the right and left footpad.
geometric mean fluorescence intensity by FACS Calibur flow cytometer
(Becton Dickinson, USA) with FL-1 filter and data were analysed by 2.14. Quantitative analysis of IgG1 and IgG2a isotypes
CellQuestPro software.
The Leishmania parasite-specific serum immunoglobulin G (IgG)
2.10. Determination of intracellular nitric oxide (NO) generation isotype antibody response was measured by a commercially available
ELISA kit (Abcam). Serum samples from each group of animals were
The level of intracellular nitric oxide (NO) in the splenocytes was added after two-fold dilutions into an antibody-coated 96-well ELISA
measured using cell permeable dye DAF-2DA. Isolated splenocytes from plate. After washing the plate HRP-labelled-secondary detector anti-
different experiment groups of animal were stimulated with SLA body (rabbit anti-mouse IgG1 or IgG2a) was added in each well. The
(50 μg/ml) for 72 h at 37 °C CO2 incubator (Thermo Electron chromogenic substrate was added and after 30 min of incubation stop

Table 2
Forward and reverse primer sequence of different genes.
Gene Sequence of forward primer Sequence of reverse primer

IL-12 5′-GGA AGC ACG GCA GCA GAA TA-3′ 5′-AAC TTG AGG GAG AAG TAG GAA TGG-3′
TNF-α 5′-CAT CTT CTC AAA ATT CGA GTG ACA A-3′ 5′-TGG GAG TAG ACA AGG TAC AAC CC-3′
IFN-γ 5′-TCA AGT GGC ATA GAT GTG GAA GAA-3′ 5′-TGG CTC TGC AGG ATT TTC ATG-3′
IL-10 5′-GGT TGC CAA GCC TTA TCG GA-3′ 5′-ACC TGC TCC ACT GCC TTG CT-3′
TGF-β 5′-TGA CGT CAC TGG AGT TGT ACG G-3′ 5′-GGT TCA TGT CAT GGA TGG TGC-3′
β-actin 5′-CGC CAT GGA TGA TGA TAT TGC-3′ 5′-AAG CCG GCC TTG CAC AT-3′

226
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Fig. 1. Determination of in vivo antileishmanial efficacy of Coccinia grandis (L.) Voigt leaf extract (Cg-Ex) on Leishmania donovani infected BALB/c mice. Estimation of
(a) splenic parasite burden and (b) hepatic parasite burden in terms of LDU in infected BALB/c mice. Cg-Ex was orally administered on L. donovani infected BALB/c
mice for consecutive 15days. Efficacy of Cg-Ex on Group I (12 mg/kg body weight), Group II (24 mg/kg body weight), Group III (48 mg/kg body weight) and SAG
treated animals (20 mg/kg body weight) was evaluated on 1day and 15days post-treatment by enumeration of intracellular parasites after Giemsa staining. (c)
Measurement of spleen weight of each group of mice. SAG (20 mg/kg body weight) was used as a positive control. Data was represented as mean ± SD (n = 3) for
three animals per group. Statistical significance was analysed between infected control and treated groups using Two-way ANOVA followed by Dunnett's multiple
comparison test (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001).

solution was added to each well. Then immediately measured the ab- 2.17. Protein estimation
sorbance on ELISA plate reader (iMark ELISA plate reader, Bio-Rad) at
450 nm with the wavelength correction at 570 nm [22]. The protein concentration of different samples was estimated by
BCA method (Pierce, Thermo Fisher Scientific).
2.15. Evaluation of biochemical markers
2.18. Densitometry analysis
For evaluation of toxicity of Cg-Ex on hepatic and renal function,
serum levels of alanine transaminase (ALT), aspartate transaminase
Densitometric analyses of semi-quantitative PCR were performed
(AST), urea and creatinine from different groups of animals were as-
with Quantity One software (Bio-Rad Laboratories, USA) and expressed
sayed using commercially available kits (Span Diagnostics Ltd., India)
as arbitrary densitometric units (ADU) in the bar plot.
[23].

2.16. Histopathological analysis 2.19. Statistical analysis

Spleen tissue fragment from each experimental set of animals was All the data were expressed as mean ± SD. Two-way ANOVA fol-
fixed in 10% neutral buffered formalin solution (Sigma-Aldrich). lowed by Dunnett's multiple comparison test was performed for statis-
Histological sections were cut into 5 μm thick after paraffin embedding tical analysis by GraphPad Prism version 8.00 (Windows, GraphPad
and mounted on microscope slides. The tissue sections were stained Software, San Diego, USA). The difference between mean of various
with haematoxylin and eosin (H&E stain, Sigma-Aldrich) and subse- groups were assessed for statistical significance, and p values < 0.05
quently examined structural changes of spleen lymphoid tissue, the cell were considered as statistically significant.
population in white pulp and red pulp under the light microscope (Axio
skop2 Plus, Zeiss) [24].

227
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Fig. 2. Quantification of intracellular reactive oxygen species (ROS) and nitric oxides (NO) in the activated splenocytes. (a) Intracellular ROS level was measured in
SLA activated splenocytes of different groups after 1day and 15days post-treatment by cell-permeable H2DCFDA dye using FACS analysis. (b) Intracellular NO was
determined in the splenocytes isolated from each group of mice after 1day and 15days post-treatment by DAF-2DA dye using FACS analysis. SAG was used as a
positive control for measurement of ROS and NO. Values are represented as the mean ± SD (n = 3), and statistical significance was examined between infected
control and treated groups by Two-way ANOVA followed by Dunnett's multiple comparison test (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001).

3. Results 65.0 ± 1.12% (LDU = 1148.5 ± 40.3) and 84.0 ± 2.1%


(LDU = 643.0 ± 21.2) with respect to the infected control at equal
3.1. Cg-Ex confers in vivo protection by reducing parasitemia time span (Fig. 1b).
Splenomegaly is one of the major hallmarks of chronic VL due to the
To validate the in vivo antileishmanial efficacy of the Cg-Ex, spleen parasitic load and cellular infiltration in the spleen [25]. With the
and liver parasite burden of experimental groups of mice were de- progression of infection spleen weight considerably increased 2.5 fold
termined in terms of LDU. Accordingly, the observation was made after and 2.7 fold at 1day and 15days post-treatment. However, the sig-
oral administration of Cg-Ex for 15days in infected mice and the nificant decrease of 1.8 fold and 2.4 fold in weight of spleen was ob-
parasite burden was measured on 1day and 15days post-treatment. In served in the Group III BALB/c mice at 1day and 15days post-treatment
case of infected control group the LDU of spleen was 755.5 ± 14.8 at with respect to the infected control mice (Fig. 1c). A similar pattern of
1day post-treatment and 834.0 ± 11.3 at 15days post-treatment. At reduction in weight of spleen was noted in SAG treated mice by 1.9 fold
lowest dose of Cg-Ex (Group I, 12 mg/kg body weight) parasite burden and 2.5 fold at 1day and 15days post-treatment respectively. Values
were decreased 29.6 ± 2.97% (LDU = 531.5 ± 16.3) and obtained from these experiments are statistically significant
44.1 ± 1.92% (LDU = 476.0 ± 9.9) from spleen at 1day and 15days (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001) compared
of post-treatment respectively. In Group II (24 mg/kg body weight) to respective control.
mice similarly the splenic parasite load was reduced by 46.2 ± 3.87%
(LDU = 406.0 ± 12.0) and 70.6 ± 5.91% (LDU = 245 ± 7.1) at
1day and 15days of post-treatment. At the highest dose of Cg-Ex (Group 3.2. Cg-Ex restores intracellular oxidative burst
III, 48 mg/kg body weight) considerable reduction of parasite burden in
the spleen has appeared with 52.2 ± 4.04% (LDU = 361.0 ± 8.5) and Intracellular Leishmania parasites deactivate the macrophages and
81.9 ± 3.84% (LDU = 151.0 ± 9.8) respectively at the same time survived by scavenging oxygen free radicals such as ROS and NO within
points. Likewise, in the positive control, i.e, SAG treated mice have the host. Nitric oxide generation depends on the activation of iNOS
shown 69.0 ± 2.67% (LDU = 234.0 ± 8.5) and 89.2 ± 1.99% expression in Leishmania infected macrophages [26]. In contrast, gen-
(LDU = 90.0 ± 7.4) reduction in splenic parasites at 1day and 15days eration of ROS and NO by macrophages upon treatment with antil-
of post-treatment respectively (Fig. 1a). This observation clearly depicts eishmanial agent facilitates the elimination of parasites from the host
that the diminution of splenic parasite burden upon Cg-Ex treatment [27]. Therefore, intracellular ROS and NO were measured in spleno-
occurs in dose-dependent manner. cytes of all groups of BALB/c mice and presented by GMFC (Geometric
Additionally, determination of liver parasite burden represents a Mean Fluorescence Channel). Splenocytes from different groups of mice
key parameter for monitoring Leishmania infection in a mouse model. In exhibited a significant elevation of intracellular ROS in response to Cg-
the infected group of mice the liver parasite burden in terms of LDU was Ex at dose-dependent manner on both time points. Splenocytes from
4139.0 ± 29.7 at 1day post-treatment and 4052.5 ± 38.9 at 15days Group III mice displayed exaggeration of ROS generation at 5.1 fold and
post treatment. Therefore, upon Cg-Ex treatment in Group I mice the 4.4 fold at 1day and 15days of post-treatment respectively when com-
diminution of liver parasite load was observed by 31.7 ± 1.01% pared to infected control. In case of other two groups (Group I and
(LDU = 2851.0 ± 26.9) and 50.6 ± 0.5% (LDU = 2039.0 ± 24.0) at Group II) ROS was also generated in dose-dependently (Fig. 2a).
1day and 15days respectively. In the same duration of time Group II The present study was also demonstrated that NO production was
BALB/c mice have exhibited 43.7 ± 0.32% (LDU = 2630.0 ± 31.1) inhibited by 2.2 fold and 3 fold in infected control compared to the
and 65.2 ± 1.47% (LDU = 1415.0 ± 37.5) decrease of parasite load uninfected animals at 1day and 15days post-treatment. Conversely,
in the liver. Correspondingly, in case of Group III treated mice liver splenocytes from Group III BALB/c mice augmented NO generation by
parasite burden was declined about 68.5 ± 1.10% 7.8 fold and 10.7 fold at 1day and 15days post-treatment in addition to
(LDU = 1269.5 ± 34.6) and 78.0 ± 2.92% (LDU = 891.5 ± 33.2) in the generation of ROS (Fig. 2b). Other two groups of Cg-Ex treated
the same time. A parallel observation was made within SAG treated animals (Group I and Group II) showed the elevation of intracellular
mice where the reduction of parasitemia from liver was about nitric oxide (NO) level in a dose-dependent manner. All data in these
two experiments are statistically significant (*p < 0.05, **p < 0.001,

228
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Fig. 3. Analysis of pro-inflammatory and anti-inflammatory cytokines secreted by stimulated splenocytes. Th1 cytokines (a) IL-12 (pg/ml), (b) TNF-α (pg/ml), (c)
IFN-γ (pg/ml) and Th2 cytokines (d) IL-10 (pg/ml), (e) TGF-β (pg/ml) were quantified in culture supernatant of splenocytes isolated from different sets of animals
after 72 h of SLA stimulation by sandwich ELISA. Results were expressed as mean ± SD (n = 3) from three replicate experiments and statistical significance was
checked using Two-way ANOVA followed by Dunnett's multiple comparison test (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001).

***p < 0.002, ****p < 0.0001) compared to the infected control. and f). Treatment with PBS in animals as vehicle control did not showed
any effect on the release of IL-12, TNF-α, and IFN-γ that proves im-
munological inertness of PBS in the in vivo model (data not shown).
3.3. Cg-Ex induces secretion of pro-inflammatory cytokines at protein and
Data are statistically significant (*p < 0.05, **p < 0.001,
mRNA level
***p < 0.002, ****p < 0.0001) compared to the respective control.

It is fully established that Leishmania infection critically skews from


pro-inflammatory response to anti-inflammatory response for sus- 3.4. Cg-Ex enhances leishmanial antigen-specific DTH response
taining the intracellular parasites in phagocytic cells [28]. Thus, the
Th1 immune response was assessed by the quantification of IL-12, TNF- DTH response predicts the peripheral blood T-cell immunity as well
α, and IFN-γ cytokine in splenocytes culture supernatant. Levels of IL- as it correlates the clearance of parasites in animal model [29]. Con-
10 and TGF-β were also used as indicators of Th2 immune response. sequently, DTH response was measured in the infected mice at 1day and
The anti-inflammatory cytokines stimulate the progression of infection 15days post-treatment. A slight increase (1.4 fold) in footpad thickness
within host. Hence, we examined whether the Cg-Ex treatment could was observed in infected mice as compared to normal mice. But a sig-
modulate pro-inflammatory cytokine (IL-12, TNF-α, and IFN-γ) and nificant rise (2.28 fold) of DTH response was observed in Group III mice
anti-inflammatory cytokine (IL-10, TGF-β) release and expression at the footpad at 15days post-treatment (Fig. 5). Each value is statistically
mRNA level in infected groups of animals. Level of Th2 cytokines were significant (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001)
found to be maximum in infected mice both at protein and mRNA level, in comparison to the infective control.
whereas their concentrations were reduced significantly in all treated
groups of animals in all post-treatment days. In Group III mice, there
was a considerable amount of increment in IL-12 (2.4 fold), TNF-α (5.6 3.5. Cg-Ex develops humoral immune responses
fold) and IFN-γ (5.8 fold) level (Fig. 3a–c) in culture supernatant of
activated splenocytes with concomitant reduction of IL-10 (3 fold) and Healing of leishmaniasis is connected with the development of
TGF-β (2.7 fold) in protein level (Fig. 3d, e). A similar effect of SAG was strong humoral immune responses like IgG2a production while estab-
observed on Th1 and Th2 cytokines in the culture supernatant at lishment of infection is associated with higher IgG1 production [30].
15days of post-treatment. The IgG2a antibody response is a marker of protective Th1 type of
The quantitative data of cytokine expression as observed by the immune response. Therefore, Cg-Ex treated BALB/c mice produced
ELISA method was confirmed by mRNA expression of the aforemen- significant level of IgG2a antibody when compared with the infected
tioned cytokines using semi-quantitative PCR analysis. The results re- control. The serum IgG2a was increased about 2 fold in Group III mice
vealed that there was a considerable inhibition of IL-12, TNF-α, and (Fig. 6a). Again, IgG1 antibody response points towards Th2 type of
IFN-γ expression in the infected splenocytes compared with uninfected immune response. Therefore, it was found that IgG1 level was higher in
splenocytes. In Cg-Ex treated infected splenocytes, 3.1, 4.0, and 3.5-fold infected BALB/c mice as compared to uninfected ones whereas IgG1
increment was shown in the expression of IL-12, TNF-α, and IFN-γ re- level was significantly reduced (2.9 fold) in the Group III mice at
spectively compared with infected splenocytes (Fig. 4a–d). On the other 15days post-treatment as compared to infected controls (Fig. 6b).
hand, mRNA profile exhibits a significant induction of IL-10 and TGF-β However, in SAG treated infected animals the increase in IgG2a level
in infected splenocytes with respect to the uninfected splenocytes. In- and decrease in level of IgG1 was lower compared with higher dose of
triguingly, when infected splenocytes were treated with Cg-Ex, there Cg-Ex treated animals (Group III). All values are statistically significant
was 3.45 and 3.62 -fold reduction in the expression of IL-10 and TGF-β (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001) compared
respectively considering with infected control splenocytes (Fig. 4a, e, to the infective control.

229
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Fig. 4. Evaluation of Th1 and Th2 cytokines in activated splenocytes using semi-quantitative PCR. The mRNA was isolated from each sample and PCR amplification
was done. PCR product of Th1 and Th2 cytokines was run on the 1.5% agarose gel (a) Lane no. 1–5 in agarose gel are represented the normal control, infected control,
Group I, Group II, and Group III of 15 days post-treatment respectively. Here, β-actin was used as control housekeeping gene. (b) The band intensities of IL-12, TNF-α,
IFN-γ, IL-10, and TGF-β were evaluated and plotted as the bar diagram. Data are represented as the mean ± SD (n = 3) and statistical significance was analysed
between infected control and treated groups by Two-way ANOVA followed by Dunnett's multiple comparison test (*p < 0.05, **p < 0.001, ***p < 0.002,
****p < 0.0001).

were done by measuring the activity of serum AST, ALT and also the
level of urea and creatinine. The activity of liver enzymes in infected
control animals have been found in higher level with respect to unin-
fected control, which was reduced in a dose dependent manner in Cg-Ex
treated infected BALB/c mice at 1day and 15days post-treatment
(Table 3). Same pattern of increment was observed in liver transami-
nase enzymes of SAG treated mice. Elevation of transaminase activity in
SAG treated mice indicates the hepatotoxicity of this conventional drug,
while reduction of these enzymes in Cg-Ex treated mice to the normal
level implies that Cg-Ex has no hepato-toxicity in mice. Values are
presented as mean ± SD and differences between means were statis-
tically significant (*p < 0.05, **p < 0.001, ***p < 0.002,
****p < 0.0001) in comparison to the infected control.
Similarly, kidney function tests also showed the normal level of urea
and creatinine in serum of uninfected mice whereas infected control
mice have higher level of urea and creatinine. Although Cg-Ex treated
mice demonstrated the significant reduction of urea and creatinine level
in the serum to normal level but during SAG treatment this level was
increased. So, these observations signify the non-nephrotoxic effect of
Cg-Ex in BALB/c mice (Table 4). Vehicle control animals showed no
toxic effect on the liver and kidney function. Normal animals treated
Fig. 5. Delayed-type hypersensitivity response induced by Cg-Ex in infected
with the highest dose of Cg-Ex (48 mg/kg body weight) did not show
BALB/c mice. DTH response was measured in various groups of BALB/c mice at
any hepatic and renal toxicity after 1day and 15days post-treatment
1day and 15days of post-treatment. Statistical significance was analysed in all
(Tables 3 and 4). Data are represented as mean ± SD and differences
groups by Two-way ANOVA followed by.
Dunnett’s multiple comparison test (*p < 0.05, **p < 0.001, ***p < 0.002, between means were statistically significant (*p < 0.05, **p < 0.001,
****p < 0.0001) and expressed as mean ± SD (n=3). ***p < 0.002, ****p < 0.0001) with respect to the infected control.

3.6. Non-toxic effect of Cg-Ex in liver and kidney function 3.7. Improvement of structural organization after Cg-Ex treatment

Hepato- and renal toxicity are common in currently available drugs Structural disorganization of white pulp and red pulp remains a
for VL [31]. In order to examine any hepatotoxicity or nephrotoxicity characteristic property of the murine model of VL [32]. Thus, histo-
exerted by Cg-Ex in the BALB/c mice, the liver and kidney function tests pathological analysis of spleen was carried out to examine the tissue
remodelling in the infected BALB/c mice. In this experiment, it was

230
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Fig. 6. Humoral immune response augmented by Cg-Ex in Leishmania donovani infected BALB/c mice. Leishmania-specific antigen (a) IgG2a and (b) IgG1 in the serum
of uninfected control, infected control and different treated groups of BALB/c mice at 1day and 15days post-treatment were quantified by the ELISA method.
Statistical significance was determined compared with infected control group (p* < 0.05) by Two-way ANOVA followed by Dunnett's multiple comparison test
(*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001) and expressed as mean ± SD (n=3).

observed that the spleen from all the groups contained an intact capsule [34,35]. However, the disease management is limited due to the small
but there was some architectural abnormality in white pulp and red therapeutic index and the emergence of resistance against available
pulp. Herein, the non-infected group of animals was shown a compact drugs for VL [36].
structure of white and red pulp (Fig. 7a) whereas the untreated infected In this perspective, searching for innovative antileishmanial agents
group has possessed extensive cellular disorganization in white and red from the natural sources is an alternative approach. Various plant ex-
pulp architecture (Fig. 7b). Interestingly, at 1day post-treatment of tracts and plant-derived products represent leishmanicidal activity by
Group III mice have showed a modest level of regeneration of white targeting different biochemical pathways [37]. Plants provide an ex-
pulp from the widespread distorted structure (Fig. 7c). At 15days of cellent source of protease inhibitors protecting from many diseases,
post-treatment, the structural integrity of white pulp and red pulp of pests, insects and herbivorous animals. Very recently scientists devoted
spleen from Group III animals seems more organized (Fig. 7d). their attention to the development of plant protease inhibitor-based
novel therapeutic agents against cardiovascular diseases, inflammatory
disease, bacterial and fungal diseases [11]. Clemente et al. have re-
4. Discussion ported that Bowman-Birk inhibitor, a serine protease inhibitor from
Pisum sativum, inhibits the in vitro growth of human colorectal adeno-
The pathogenesis of visceral leishmaniasis concerns about the sub- carcinoma cells [38]. Again, the serine protease inhibitor of Coccinia
version and modulation of both innate and adaptive immunity of the grandis demonstrated the highest inhibitory activity against trypsin and
host leading to immunosuppression in individuals [33]. Hence, the chymotrypsin as well as exhibiting the antimicrobial and antifungal
prevention of VL is challenging and the initiation of an appropriate activity [13]. It is also noted that some plant-derived protease inhibitor
immune response is a necessary criterion to cure the disease. In fact, has shown antileishmanial activity with non-toxic effect on the mac-
conventional drugs like sodium stibogluconate and miltefosine act as an rophages [5]. In another report, epoxy-α-lapachone, a plant-derived
immunomodulator that elicits the Th1-biased response and activates serine protease inhibitor of oligopeptidase B, was also found to reduce
the oxidative burst in macrophages to eliminate Leishmania parasites

Table 3
Effect of Cg-Ex on liver enzymes activity of different groups of BALB/c mice.
Enzymes Post Normal control Normal Infected control Vehicle Group I Group II Group III (48mg/ Sodium
(Normal treatment control+ control (12mg/kg §b.w (24mg/kg §b.w kg §b.w †Cg-Ex) stibogluconate
† †
range) days 48mg/kg §b.w Cg-Ex) Cg-Ex) (20mg/kg §b.w)

Cg-Ex

AST 1 22.36 ± 3.11 23.17 ± 2.34 50.97 ± 1.72 51.26 ± 1.45 38.51 ± 0.88 33.06 ± 2.27 27.61 ± 2.46 61.90 ± 2.36
(5-40 (*p˂0.05) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)
IU/L)
15 26.92 ± 2.32 24.56 ± 2.78 59.43 ± 2.72 60.14 ± 2.56 31.79 ± 1.99 27.64 ± 2.08 24.96 ± 3.98 67.20 ± 2.94
(****p˂0.0001) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)

#
ALT 1 24.31 ± 2.61 24.11 ± 1.31 60.61 ± 2.44 59.97 ± 2.26 54.69 ± 2.99 33.21 ± 1.32 30.95 ± 2.42 76.96 ± 1.86
(5-35 (****p˂0.0001) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)
IU/L)

15 26.24 ± 2.81 25.67 ± 2.45 75.31 ± 2.94 77.37 ± 1.82 46.56 ± 2.05 31.27 ± 1.69 27.05 ± 1.40 82.96 ± 2.57
(****p˂0.0001) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)

#
ALT = Alanine Transaminase; AST = Aspartate Transaminase, b.w = body weight, Cg-Ex=Coccinia grandis (L.) Voigt leaf extract, SAG = Sodium stibogluconate.
†BALB/c mice (n = 3) received Cg-Ex for consecutive 15days and activity of liver enzymes were estimated after 1day and 15days post-treatment (mean ± SD) using
commercial kits. All values are statistically significant (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001) and statistical significance was analysed with
respect to the infected control using Two-way ANOVA followed by Dunnett's multiple comparison test.

231
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Table 4
Consequence of Cg-Ex on the renal function of various groups of BALB/c mice.
Metabolites Post Normal control (Normal Infected control Vehicle Group I Group II (24mg/ Group III (48mg/ Sodium
(Normal treatment control+ control (12mg/kg kg b.w†Cg-Ex) kg b.w†Cg-Ex) stibogluconate
range) days 48mg/ b.w†Cg-Ex) (20mg/kg b.w)
kg§b.w†Cg-Ex)

Urea 1 32.98 ± 1.74 33.21 ± 2.08 57.63 ± 3.32 55.12 ± 2.67 49.06 ± 1.34 40.57 ± 1.87 33.86 ± 1.97 68.58 ± 1.29
(10-45 (**p˂0.001) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)
mg/dl)
15 36.57 ± 2.18 35.56 ± 2.64 65.12 ± 1.49 66.78 ± 2.23 43.33 ± 1.43 34.09 ± 1.72 31.84 ± 1.69 80.46 ± 2.05
(****p˂0.0001) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)

Creatinine 1 0.87 ± 0.03 0.86 ± 0.02 1.45 ± 0.06 1.49 ± 0.02 1.35 ± 0.04 1.20 ± 0.01 0.92 ± 0.04 1.54 ± 0.03
(0.6-1.2 (*p˂0.05) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)
mg%)
15 0.85 ± 0.03 0.85 ± 0.04 1.68 ± 0.04 1.72 ± 0.07 1.27 ± 0.01 1.14 ± 0.04 0.82 ± 0.02 1.88 ± 0.02
(****p˂0.0001) (****p˂0.0001) (****p˂0.0001) (****p˂0.0001)

#
b.w = body weight, Cg-Ex=Coccinia grandis (L.) Voigt leaf extract, SAG = Sodium stibogluconate.
†Cg-Ex was orally administered in BALB/c mice (n = 3) for consecutive 15days and metabolites were estimated after 1day and 15days post-treatment (mean ± SD)
by commercial kits. All values are statistically significant (*p < 0.05, **p < 0.001, ***p < 0.002, ****p < 0.0001) and statistical significance of different groups
were examined with respect to the infected control group using Two-way ANOVA followed by Dunnett's multiple comparison test.

Fig. 7. Assessment of histopathological alterations in the spleen after treatment of Cg-Ex.


Histology of spleen of BALB/c mice (a) uninfected control (b) infected control (c) Cg-Ex treated Group III mice (48 mg/kg body weight) at 1day post-treatment (d) Cg-
Ex treated Group III mice (48 mg/kg body weight) at 15days post-treatment. Tissue sections were stained with H&E stain and scale bar is 20 μm (20X magnification).
Capsule, white pulp, and red pulp are indicated by the black arrow in the histological section of spleen. (For interpretation of the references to colour in this figure
legend, the reader is referred to the web version of this article.)

232
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

lesion size in Leishmania amazonensis infected BALB/c mice [39]. Ad- decreases IgG1 level in treated BALB/c mice [45]. Therefore, con-
ditionally, our previous reports have shown the antileishmanial activity sidering these observations it can be concluded that Cg-Ex activates
of Coccinia grandis (L.) Voigt leaf extract (Cg-Ex) that is enriched with both cell-mediated immunity and humoral immunity to clear the
three serine protease inhibitors (64.8, 55.8 and 15.3 kDa). The Leish- parasitic burden from infected mice.
manial intracellular serine protease inhibitor (58 kDa, LD-SP) was in- Hepatomegaly is a characteristic property of the VL and renal dys-
hibited by Cg-Ex without displaying any toxicity on murine macro- functions have been well documented that comprises interstitial and
phages [14,15]. The present study revealed that Cg-Ex exerts glomerular defect [46]. Furthermore, the renal and hepatic dysfunction
antileishmanial activity on mouse model of VL by reducing the parasite are major side effects of the currently available drugs used for treatment
growth. Another investigation stated that the leaf extract of Croton of VL [31]. A significant decrease of transaminase enzyme activity and
caudatus eliminates in vivo parasitic burden by up regulation of Th1 serum urea and creatinine level indicate the non-toxic effect of Cg-Ex on
cells of the host [40]. Thus, other reports support our present findings liver and kidney. Similarly, the plant derived bioactive fraction of Piper
of antileishmanial activity of Cg-Ex on murine model of VL. nigum has demonstrated the in vivo nontoxic effect on liver and kidney
For the establishment of infection within host macrophages, function with antileishmanial property [47]. Here, the SAG treated
Leishmania parasites hinder the production of parasiticidal molecules animals showed antileishmanial activity with extensive toxicity in liver
from macrophages such as reactive oxygen species (ROS) and reactive and kidney whereas Cg-Ex does not express any hepatotoxicity and
nitrogen species (RNS). Therefore, macrophage-derived reactive inter- nephrotoxicity in the animal model of VL.
mediates are involved in the innate host defence mechanism as integral During visceral leishmaniasis, extensive damage of tissue occurs in
components for controlling Leishmania infection within the host cell the spleen due to the disruption of both fibroblastic reticular cell net-
[27]. It is familiar that IFN-γ stimulates the expression of iNOS with work and follicular dendritic cell network [28]. Upon oral adminis-
substantial production of nitric oxide in macrophages [41]. The present tration of Cg-Ex, the cellular disorganization in white pulp and red pulp
study demonstrated that Cg-Ex exerts antileishmanial activity through of spleen was restored by preventing tissue disruption in the infected
the production of ROS and NO in infected BALB/c mice. Our observa- BALB/c mice. Dalton et al. demonstrated that antimonial drug along
tions very clearly depicted that Cg-Ex could effectively mediate NO with sunitinib maleate (Sm), an inhibitor of receptor tyrosine kinase,
generation as it significantly induced iNOS expression at the protein enhanced leishmanicidal activity as well as block tissue remodelling
level in L. donovani-infected animals. The similar effect was shown by and splenomegaly in experimental VL [48]. In the similar fashion, ur-
cystatin, a natural cysteine protease inhibitor, which exhibits in vivo solic acid, a plant-derived compound, showed the reduction of parasitic
leishmanicidal effect by switching Th2 to Th1 cytokine responses and burden in liver and spleen as well as changed the histological archi-
also produced intracellular NO via the activation of JAK/STAT pathway tecture of spleen white pulp [49].
[42]. Our overall findings indicate that Cg-Ex possesses a protective role
The chemotherapeutic efficacy of a drug for VL includes restoration in the L. donovani infected BALB/c mice model without nephrotoxicity
of T-cell response as this response is impaired during the progression of and hepatotoxicity after oral administration. The potential antil-
the disease. Macrophages and T-cell subsets secret IL-12 that helps in eishmanial activity of Cg-Ex was augmented by both cell mediated and
the development of Th1 cells and the effectiveness of antileishmanial humoral immunity of the host as described in Fig. 8 and Cg-Ex also
therapy is related with the reestablishment of IFN-γ and IL-12 secretion prevents the tissue damage in the spleen. Consequently, Cg-Ex can be a
[33]. deliberate candidate for achieving a new identity as therapeutic agent
Moreover, Leishmania proteases particularly cysteine proteases and against visceral leishmaniasis.
gp63 play a crucial role in the immune evasion process by degradation
of various proteins of host, such as the complement factors (C3b), 5. Conclusions
transcription factor NF-κB and the MHC class II molecules which
modify the immune response of the macrophages and facilitates per- Altogether, our study illustrates the in vivo leishmanicidal potency
sistence of infection within the host. The pieces of literature also re- of Cg-Ex that occurs by means of the up regulation of Th1 immune
ported that some natural protease inhibitors have shown leishmanicidal response and simultaneous production of ROS and NO exerting non-
activity by inhibiting the leishmania proteases and also stimulates the toxic effect on the mouse model. Combining results of our studies as
Th1 responses in macrophages [4,5,26]. The oral administration of Cg- observed previously in vitro and currently in vivo systems, it is suggested
Ex has also enhanced Th1 response especially IL-12, TNF-α, and IFN-γ that serine protease inhibitor enriched leaf extract (Cg-Ex) can inhibit
with simultaneous diminish of IL-10 and TGF-β in the infected BALB/c the Leishmania donovani serine protease and could be a potential option
mice. Several reports suggested that various plant-derived products for VL chemotherapy. The current study accredits further investigations
exert antileishmanial activity via modulation of T-cell response [43]. to identify the Cg-Ex-derived highly active protease inhibitor against
Delayed-type hypersensitivity (DTH) or type-IV cell-mediated hy- Leishmania serine protease. This endeavour might facilitate the ex-
persensitivity represents a localized inflammatory reaction where Th1 ploration of the exact mechanisms behind the antileishmanial activity
cells play a pivotal role in this response. DTH response induces the of a novel natural plant derived lead component in the treatment of VL.
clearance of parasites from infected host cells by activation of T-cell
that leads to the release of Th1 cytokines resulting in the infiltration of
lymphocytes and macrophages into the infected tissue [44]. It is re- Conflict of interest
ported that plant-derived immunomodulator, eugenol exhibits DTH
response in BALB/c mice. This observation suggested that higher the The authors declare that they have no conflict of interest.
DTH response lower the parasitic load and greater the efficacy of im-
munomodulatory plant-derived compound [23]. The current study re- Acknowledgements
ported that DTH response was induced by Cg-Ex in BALB/c mice.
During the course of Leishmania infection, the host IgG1 increases AP is recipient of ICMR-SRF and this experimental work was fi-
which is responsible for secretion of disease-promoting IL-10 cytokine nancially supported by the grants from the Indian Council of Medical
whereas IgG2a antibody decreases after resolution of infection that is Research (ICMR, Government of India), New Delhi (sanction No.
linked with IFN-γ production [30]. Thus, the detail immunological Fellowship/18/2018-ECD-II), Government of India. Authors are
study demonstrated that Cg-Ex treatment induces production of IgG2a thankful to the DST-FIST phase-III (Project no. FIST Sl. NO. 185),
level due to the up-regulation of IFN-γ. Azadirachta indica bioactive Government of India and UGC-SAP phase-II (Project No. F 5-3/2018/
fraction significantly increases serum IgG2a level but simultaneously DRS-II) and University of Kalyani.

233
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Fig. 8. Summary of immunological responses during establishment of infection and elimination of infection from Leishmania donovani infected BALB/c mice upon
oral administration of Cg-Ex. The figure illustrates that during L. donovani infection in BALB/c mice, the Th2 type immune response was predominant with sup-
pression of ROS and NO production. In contrast, Th1 type immune response is up-regulated following treatment with Cg-Ex. The generation of ROS and NO with
positive DTH response facilitates reduction of the parasite burden in the infected BALB/c mice.

References [15] A. Pramanik, D. Paik, K. Naskar, T. Chakraborti, Coccinia grandis (L.) Voigt leaf
extract exhibits antileishmanial effect through pro-inflammatory response: an in
vitro study, Curr. Microbiol. 74 (2017) 59–67, https://doi.org/10.1007/s00284-
[1] M. Ghorbani, R. Farhoudi, Leishmaniasis in humans: drug or vaccine therapy? Drug 016-1151-4.
Des. Dev. Ther. 12 (2017) 25–40, https://doi.org/10.2147/DDDT.S146521. [16] J. Paul, S. Karmakar, T. De, TLR-mediated distinct IFN-γ/IL-10 pattern induces
[2] S. Sundar, A. Singh, Chemotherapeutics of visceral leishmaniasis: present and future protective immunity against murine visceral leishmaniasis, Eur. J. Immunol. 42
developments, Parasitology 145 (2018) 481–489, https://doi.org/10.1017/ (2012) 2087–2099, https://doi.org/10.1002/eji.201242428.
S0031182017002116. [17] S.K. Bhaumik, J. Paul, K. Naskar, S. Karmakar, T. De, Asiaticoside induces tumour-
[3] A.S. Nagle, S. Khare, A.B. Kumar, F. Supek, A. Buchynskyy, C.J. Mathison, necrosis-factor-α-mediated nitric oxide production to cure experimental visceral
N.K. Chennamaneni, N. Pendem, F.S. Buckner, M.H. Gelb, V. Molteni, Recent de- leishmaniasis caused by antimony-susceptible and -resistant Leishmania donovani
velopments in drug discovery for leishmaniasis and human African trypanoso- strains, J. Antimicrob. Chemother. 67 (2012) 910–920, https://doi.org/10.1093/
miasis, Chem. Rev. 114 (2014) 11305–11347, https://doi.org/10.1021/cr500365f. jac/dkr575.
[4] M. Olivier, K. Hassani, Protease inhibitors as prophylaxis against leishmaniasis: new [18] S.K. Bhaumik, K. Naskar, T. De, Complete protection against experimental visceral
hope from the major surface protease gp63, Future Med. Chem. 2 (2010) 539–542. leishmaniasis with complete soluble antigen from attenuated Leishmania donovani
[5] C.R. Alves, R.S. Souza, K.D.S. Charret, L.M.C. Côrtes, M.P. Sá-Silva, L. Barral-Veloso, promastigotes involves Th1-immunity and down-regulation of IL-10, Eur. J.
L.F.G. Oliveira, F.S. da Silva, Understanding serine proteases implications on Immunol. 39 (2009) 2146–2160, https://doi.org/10.1002/eji.200839017.
Leishmania spp. lifecycle, Exp. Parasitol. 184 (2018) 67–81, https://doi.org/10. [19] S. Kaur, T. Kaur, N. Garg, S. Mukherjee, P. Raina, V. Athokpam, Effect of dose and
1016/j.exppara.2017.11.008. route of inoculation on the generation of CD4+ Th1/Th2 type of immune response
[6] R. Choudhury, S.K. Bhaumik, T. De, T. Chakraborti, Identification, purification, and in murine visceral leishmaniasis, Parasitol. Res. 103 (2008) 1413–1419, https://
characterization of a secretory serine protease in an Indian strain of Leishmania doi.org/10.1007/s00436-008-1150-x.
donovani, Mol. Cell. Biochem. 320 (2009) 1–14, https://doi.org/10.1007/s11010- [20] L.A. Stauber, Host resistance to Khartoum strain of Leishmania donovani, Rice Inst.
008-9849-7. Pamphlet 45 (1958) 80–83.
[7] R. Choudhury, P. Das, S.K. Bhaumik, T. De, T. Chakraborti, In situ im- [21] A. Sarkar, P. Saha, G. Mandal, D. Mukhopadhyay, S. Roy, S.K. Singh, S. Das,
munolocalization and stage-dependent expression of a secretory serine protease in R.P. Goswami, B. Saha, D. Kumar, P. Das, M. Chatterjee, Monitoring of intracellular
Leishmania donovani and its role as a vaccine candidate, Clin. Vaccine Immunol. 17 nitric oxide in leishmaniasis: its applicability in patients with visceral leishmaniasis,
(2010) 660–667, https://doi.org/10.1128/CVI.00358-09. Cytometry A 79 (2011) 35–45, https://doi.org/10.1002/cyto.a.21001.
[8] R. Choudhury, P. Das, T. De, T. Chakraborti, Immunolocalization and character- [22] H. Sachdeva, R. Sehgal, S. Kaur, Tinospora cordifolia as a protective and im-
ization of two novel proteases in Leishmania donovani: putative roles in host in- munomodulatory agent in combination with cisplatin against murine visceral
vasion and parasite development, Biochimie 92 (2010) 1274–1286, https://doi. leishmaniasis, Exp. Parasitol. 137 (2014) 53–65, https://doi.org/10.1016/j.
org/10.1016/j.biochi.2010.05.015. exppara.2013.12.006.
[9] G.M. Cragg, D.J. Newman, Natural products: a continuing source of novel drug [23] M. Islamuddin, G. Chouhan, M.Y. Want, H.A. Ozbak, H.A. Hemeg, F. Afrin,
leads, Biochim. Biophys. Acta 1830 (2013) 3670–3695, https://doi.org/10.1016/j. Immunotherapeutic potential of eugenol emulsion in experimental visceral leish-
bbagen.2013.02.008. maniasis, PLoS Negl. Trop. Dis. 10 (2016) e0005011, https://doi.org/10.1371/
[10] D.A. Dias, S. Urban, U. Roessner, A historical overview of natural products in drug journal.pntd.0005011.
discovery, Metabolites 2 (2012) 303–336, https://doi.org/10.3390/ [24] P. Yurdakul, J. Dalton, L. Beattie, N. Brown, S. Erguven, A. Maroof, P.M. Kaye,
metabo2020303. Compartment-specific remodeling of splenic micro-architecture during experi-
[11] S. Srikanth, Z. Chen, Plant protease inhibitors in therapeutics-focus on cancer mental visceral leishmaniasis, Am. J. Pathol. 179 (2011) 23–29, https://doi.org/10.
therapy, Front. Pharmacol. 7 (2016) 470, https://doi.org/10.3389/fphar.2016. 1016/j.ajpath.2011.03.009.
00470. [25] P.M. Kaye, M. Svensson, M. Ato, A. Maroof, R. Polley, S. Stager, S. Zubairi,
[12] N. Tamilselvan, T. Thirumalai, E.K. Elumalai, R. Balaji, E. David, Pharmacognosy of C.R. Engwerda, The immunopathology of experimental visceral leishmaniasis,
Coccinia grandis: a review, Asian Pac. J. Trop. Biomed. 1 (2011) 299–302. Immunol. Rev. 201 (2004) 239–253.
[13] L.S. Satheesh, K. Murugan, Antimicrobial activity of protease inhibitor from leaves [26] M. Olivier, D.J. Gregory, G. Forget, Subversion mechanisms by which Leishmania
of Coccinia grandis (L.) Voigt, Indian J. Exp. Biol. 49 (2011) 366–374, https://doi. parasites can escape the host immune response: a signaling point of view, Clin.
org/10.1007/s00284-016-1151-1154. Microbiol. Rev. 18 (2005) 293–305, https://doi.org/10.1128/CMR.18.2.293-305.
[14] P. Das, D. Paik, A. Pramanik, T. De, T. Chakraborti, Antiproteolytic and leishma- 2005.
nicidal activity of Coccinia grandis (L.) Voigt leaf extract against Leishmania dono- [27] H.W. Murray, C.F. Nathan, Macrophage microbicidal mechanisms in vivo: reactive
vani promastigotes, Indian J. Exp. Biol. 53 (2015) 740–746. nitrogen versus oxygen intermediates in the killing of intracellular visceral

234
A. Pramanik et al. Biomedicine & Pharmacotherapy 111 (2019) 224–235

Leishmania donovani, J. Exp. Med. 189 (1999) 741–746. this parasite, Antimicrob. Agents Chemother. 59 (2015) 1910–1918, https://doi.
[28] A.C. Stanley, C.R. Engwerda, Balancing immunity and pathology in visceral leish- org/10.1128/AAC.04742-14.
maniasis, Immunol. Cell Biol. 85 (2007) 138–147. [40] S. Dey, D. Mukherjee, S. Chakraborty, S. Mallick, A. Dutta, J. Ghosh, N. Swapana,
[29] A.R. Khabiri, F. Bagheri, M. Assmar, Leishmania major: common antigen re- S. Maiti, N. Ghorai, C.B. Singh, C. Pal, Protective effect of Croton caudatus Geisel
sponsible for induction of delayed-type hypersensitivity response in guinea pigs, leaf extract against experimental visceral leishmaniasis induces proinflammatory
Parasitol. Res. 100 (2007) 629–632. cytokines in vitro and in vivo, Exp. Parasitol. 151–152 (2015) 84–95, https://doi.
[30] R. Basu, S. Bhaumik, J.M. Basu, K. Naskar, T. De, S. Roy, Kinetoplastid membrane org/10.1016/j.exppara.2015.01.012.
protein-11 DNA vaccination induces complete protection against both pentavalent [41] T.I. Roach, A.F. Kiderlen, J.M. Blackwell, Role of inorganic nitrogen oxides and
antimonial-sensitive and -resistant strains of Leishmania donovani that correlates tumor necrosis factor alpha in killing Leishmania donovani amastigotes in gamma
with inducible nitric oxide synthase activity and IL-4 generation: evidence for interferon-lipopolysaccharide-activated macrophages from Lshs and Lshr congenic
mixed Th1- and Th2-like responses in visceral leishmaniasis, J. Immunol. 174 mouse strains, Infect. Immun. 59 (1991) 3935–3944.
(2005) 7160–7171. [42] S. Mukherjee, A. Ukil, P.K. Das, Immunomodulatory peptide from cystatin, a natural
[31] S. Sundar, J. Chakravarty, An update on pharmacotherapy for leishmaniasis, Expert cysteine protease inhibitor, against leishmaniasis as a model macrophage disease,
Opin. Pharmacother. 16 (2015) 237–252, https://doi.org/10.1517/14656566. Antimicrob. Agents Chemother. 51 (2007) 1700–1707.
2015.973850. [43] G. Chouhan, M. Islamuddin, D. Sahal, F. Afrin, Exploring the role of medicinal
[32] A.S. Cavalcanti, M. Ribeiro-Alves, O. Pereira Lde, G.L. Mestre, A.B. Ferreira, plant-based immunomodulators for effective therapy of leishmaniasis, Front.
F.N. Morgado, M.C. Boité, E. Cupolillo, M.O. Moraes, R. Porrozzi, Parasite load Immunol. 5 (2014) 193, https://doi.org/10.3389/fimmu.2014.00193.
induces progressive spleen architecture breakage and impairs cytokine mRNA ex- [44] A. Lechner, U. Ritter, R. Varona, G. Marquez, C. Bogdan, H. Körner, Protective
pression in Leishmania infantum-naturally infected dogs, PLoS One 10 (2015) immunity and delayed type hypersensitivity reaction are uncoupled in experimental
e0123009, https://doi.org/10.1371/journal.pone.0123009. Leishmania major infection of CCR6-negative mice, Microbes Infect. 9 (2007)
[33] R. Kumar, S. Nylen, Immunobiology of visceral leishmaniasis, Front. Immunol. 3 291–299.
(2012) 251, https://doi.org/10.3389/fimmu.2012.00251. [45] G. Chouhan, M. Islamuddin, M.Y. Want, M.Z. Abdin, H.A. Ozbak, H.A. Hemeg,
[34] K. Fan, E. Borden, T. Yi, Interferon-gamma is induced in human peripheral blood D. Sahal, F. Afrin, Apoptosis mediated leishmanicidal activity of Azadirachta indica
immune cells in vitro by sodium stibogluconate/interleukin-2 and mediates its bioactive fractions is accompanied by Th1 immunostimulatory potential and ther-
antitumor activity in vivo, J. Interferon Cytokine Res. 29 (2009) 451–460, https:// apeutic cure in vivo, Parasites Vectors 8 (2015) 183, https://doi.org/10.1186/
doi.org/10.1089/jir.2008.0061. s13071-015-0788-3.
[35] P. Wadhone, M. Maiti, R. Agarwal, V. Kamat, S. Martin, B. Saha, Miltefosine pro- [46] M.J. Oliveira, G.B. Silva Júnior, K.L. Abreu, N.A. Rocha, A.V. Garcia, L.F. Franco,
motes IFN-gamma-dominated anti-leishmanial immune response, J. Immunol. 182 R.M. Mota, A.B. Libório, E.F. Daher, Risk factors for acute kidney injury in visceral
(2009) 7146–7154, https://doi.org/10.4049/jimmunol.0803859. leishmaniasis (Kala-Azar), Am. J. Trop. Med. Hyg. 82 (2010) 449–453, https://doi.
[36] O.P. Singh, S. Sundar, Immunotherapy and targeted therapies in treatment of org/10.4269/ajtmh.2010.09-0571.
visceral leishmaniasis: current status and future prospects, Front. Immunol. 5 [47] G. Chouhan, M. Islamuddin, M.Y. Want, H.A. Ozbak, H.A. Hemeg, D. Sahal, F. Afrin,
(2014) 296, https://doi.org/10.3389/fimmu.2014.00296. Leishmanicidal activity of Piper nigrum bioactive fractions is interceded via
[37] R. Sen, M. Chatterjee, Plant derived therapeutics for the treatment of Leishmaniasis, apoptosis in vitro and substantiated by Th1 immunostimulatory potential in vivo,
Phytomedicine 18 (2011) 1056–1069, https://doi.org/10.1016/j.phymed.2011.03. Front. Microbiol. 6 (2016) 1368, https://doi.org/10.3389/fmicb.2015.01368.
004. [48] J.E. Dalton, A. Maroof, B.M. Owens, P. Narang, K. Johnson, N. Brown,
[38] A. Clemente, M. Carmen Marín-Manzano, E. Jiménez, M. Carmen Arques, L. Rosenquist, L. Beattie, M. Coles, P.M. Kaye, Inhibition of receptor tyrosine ki-
C. Domoney, The anti-proliferative effect of TI1B, a major Bowman-Birk iso- nases restores immunocompetence and improves immune-dependent chemotherapy
inhibitor from pea (Pisum sativum L.), on HT29 colon cancer Cells is mediated against experimental leishmaniasis in mice, J. Clin. Invest. 120 (2010) 1204–1216,
through protease inhibition, Br. J. Nutr. 1 (2012) S135–S144, https://doi.org/10. https://doi.org/10.1172/JCI41281A.
1017/S000711451200075X. [49] J.A. Jesus, T.N. Fragoso, E.S. Yamamoto, M.D. Laurenti, M.S. Silva, A.F. Ferreira,
[39] F. Souza-Silva, S.C. Bourguignon, B.A. Pereira, L.M. Côrtes, L.F. de Oliveira, J.H. Lago, G. Santos-Gomes, L.F. Passero, Therapeutic effect of ursolic acid in ex-
A. Henriques-Pons, L.C. Finkelstein, V.F. Ferreira, P.F. Carneiro, R.T. de Pinho, perimental visceral leishmaniasis, Int. J. Parasitol. Drugs Drug Resist. 7 (2017)
E.R. Caffarena, C.R. Alves, Epoxy-α-lapachone has in vitro and in vivo anti-leish- 1–11, https://doi.org/10.1016/j.ijpddr.2016.12.002.
mania (Leishmania) amazonensis effects and inhibits serine proteinase activity in

235

You might also like