Sunteți pe pagina 1din 25

Accepted Manuscript

Monoterpenoid perillyl alcohol impairs metabolic flexibility of Candida albicans by


inhibiting glyoxylate cycle

Moiz A. Ansari, Zeeshan Fatima, Kamal Ahmad, Saif Hameed

PII: S0006-291X(17)32242-8
DOI: 10.1016/j.bbrc.2017.11.064
Reference: YBBRC 38857

To appear in: Biochemical and Biophysical Research Communications

Received Date: 6 November 2017

Accepted Date: 8 November 2017

Please cite this article as: M.A. Ansari, Z. Fatima, K. Ahmad, S. Hameed, Monoterpenoid perillyl
alcohol impairs metabolic flexibility of Candida albicans by inhibiting glyoxylate cycle, Biochemical and
Biophysical Research Communications (2017), doi: 10.1016/j.bbrc.2017.11.064.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT

Mitochondria membrane Ψ
Ansari et al. 2017,
ADMET DMPK

PT
Na+/K+ transport

RI
Ansari et al. 2016,
PLoS One Cell wall Cell membrane Virulence traits

U SC
Antifungal effects of Perillyl alcohol

AN
M
Present study
In vivo efficacy of

D
perillyl alcohol
Malate TE Isocitrate
EP
Enhanced C. elegans
survival
Glyoxylate
C
AC

Glyoxylate Cycle Less hemolytic activity


inhibition
ACCEPTED MANUSCRIPT

Monoterpenoid perillyl alcohol impairs metabolic flexibility of Candida albicans by

inhibiting glyoxylate cycle

Moiz A. Ansari1#, Zeeshan Fatima1#, Kamal Ahmad2 and Saif Hameed1*

PT
1
Amity Institute of Biotechnology, Amity University Haryana, Gurugram (Manesar)-122413,
India. 2Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi-
110025, India.

RI
SC
Running Title: Perillyl alcohol inhibits glyoxylate cycle

U
AN
Abbreviations: Perillyl alcohol (PA), Isocitrate lyase (ICL), malate synthase (MLS), Glyoxylate
M

cycle (GC)
D
TE

*Corresponding Author: Dr. Saif Hameed, Amity Institute of Biotechnology, Amity University

Haryana, Gurugram (Manesar)-122413, India. Phone: +91-124-2337015, Ext: 1116.


EP

Email: saifhameed@yahoo.co.in, shameed@ggn.amity.edu


C
AC

1
ACCEPTED MANUSCRIPT

Abstract

The metabolic pathway such as glyoxylate cycle (GC) enables Candida albicans, to survive

under glucose deficient conditions prevalent in the hostile niche. Thus its key enzymes (Isocitrate

PT
lyase; ICL and malate synthase; MLS) represent attractive targets against C. albicans. We have

previously reported the antifungal potential of a natural monoterpenoid perillyl alcohol (PA).

RI
The present study uncovers additional role of PA as a potent GC inhibitor. We explored that PA

SC
phenocopied ICL1 deletion mutant and were hypersensitive under low carbon utilizing

conditions. The effect of PA on GC was substantiated by molecular docking analyses, which

U
reveals the in-silico binding affinity of PA with ICL and MLS and explored that PA binds to the
AN
active sites of both proteins with better binding energy in comparison to their known inhibitors

3-nitropropionate and bromopyruvate respectively. Enzyme kinetics by Lineweaver-Burk plot


M

unravels that PA inhibits ICL and MLS enzymes in competitive and non-competitive manner

respectively. Moreover, semi-quantitative RT-PCR indicated that PA inhibits ICL1 and MLS1
D

mRNA expressions. Lastly, we demonstrated the antifungal efficacy of PA by enhanced survival


TE

of Caenorhabditis elegans model and less hemolytic activity (29%) on human blood cells.

Further studies are warranted for PA to be considered as viable drug candidate.


C EP

Key words: perillyl alcohol; Candida; glyoxylate cycle; isocitrate lyase; malate synthase;
AC

virulence;

2
ACCEPTED MANUSCRIPT

Introduction

Candida albicans is a major source of fungal infection and leading cause of nosocomial

infections particularly under immunocompromised conditions such as HIV/AIDS, cancer,

PT
transplantations, neonatal infections etc [1,2]. With the impeding progress in the development of

new antifungal drugs and concomitant rise in multidrug resistance against existing drugs this

RI
problem has augmented [3,4,5]. Phytochemicals such as terpenoids, alkaloids, tannins,

SC
flavonoids, owing to their rich diversity and subordinate toxicity have become renewed source of

interest nowadays and been widely studied to be exploited for their antifungal potential [6].

U
Perillyl alcohol (PA) is one such natural monoterpenoid which is known for having promising
AN
antifungal properties [7,8].

Although glucose serves as the key source of carbon for C. albicans but most often the fungus
M

encounters environmental niches where low carbon compounds such as acetate, citrate and fatty

acids are prevalent which C. albicans must surmount to trigger the virulence [9,10]. Thus
D

metabolic flexibility is one factor which is crucial for C. albicans to establish successful
TE

infection as a strategy to prevail the hostile niche having nutrient limited conditions. In

microorganisms, glyoxylate cycle (GC) acts as a significant metabolic bypass for tricarboxylic
EP

acid cycle (TCA cycle) to consume simple carbon (C2) compounds in a two step process when
C

low amount of glucose is available as a carbon source. This cycle thus permits the use of C2
AC

compounds and prevents the loss of two carbons by bypassing the steps of CO2 generation in

TCA cycle and allows anaplerotic maintenance of TCA cycle intermediates [11,12]. This cycle

consist of five enzymes including isocitrate lyase (ICL) and malate synthase (MLS) encoded by

ICL1 and MLS1 respectively which are unique and can act as antifungal target as they do not

exists in humans [13]. Furthermore, C. albicans which lacks either of ICL1 and MLS1 are less

3
ACCEPTED MANUSCRIPT

virulent in mouse models of systemic candidiasis and less persistent than the wild type strains

thereby proving its indispensability to establish successful infection [14,15]. Thus the aim of the

present study was to elucidate the potential of PA as inhibitor of C. albicans GC enzymes and

demonstrate the in vivo antifungal efficacy with C. elegans as a model organism.

PT
RI
Materials and methods

SC
Candida strains and media

The C. albicans strains SC5314 was used as wild type along with GC mutants, MRC10

U
(∆icl1/icl1) and MRC11 (∆icl1+ICL1) [16]. Strains were grown in yeast nitrogen base (YNB)
AN
contained 0.67% YNB and 2% agar (for solid plates) supplemented with different carbon sources
M

either 2% glucose, 2% citrate, 2% acetate, 5% ethanol and 2% glycerol. To check the persistence

of Candida cells in C. elegans, Act1p-GFP [17] was grown on brain heart infusion (BHI) media
D

and was fed to C. elegans.


TE

Drop dilution test

To check the phenotype susceptibility under glucose deprived conditions drop dilution assay was
EP

performed as described previously in the absence (control) and presence of PA [7]. Briefly, 5µl

of fivefold serially diluted yeast cultures (OD600 0.1) were spotted onto YEPD plates. Growth
C

difference was measured after 48 hours at 30°C.


AC

3D structure prediction by I-TASSER and validation

The 550 and 551 amino acid sequence of ICL and MLS respectively of C. albicans was retrieved

from Swiss Prot-database in FASTA format. I-TASSER server, to generate 3D model of query

4
ACCEPTED MANUSCRIPT

sequence by multiple threading alignments and iterative structural assembly simulation, was used

to generate 3D model of both proteins [18].

Molecular docking

AutoDock 4.2 with standard protocol was used to dock the ICL and MLS inhibitor and PA into

PT
the active site of ICL and MLS [19,20]. The energy calculations were done using Lamarckian

RI
genetic algorithm (LGA). The conformations with the most favorable free binding energy were

selected for analyzing the interactions between the target receptor and ligands by PyMOL [19].

SC
ICL enzyme assay

Candida cells were grown in the YNBL media supplemented with 2% acetate to trigger the

U
expression of GC enzymes. The cells were harvested by centrifugation at 5,000xg for 3 minutes.
AN
Cell-free extract was prepared as described elsewhere [21], with some modifications. Briefly, the

cells were washed once with lysis buffer (100 mM potassium phosphate buffer, pH 7.5; 2 mM
M

MgCl2; 1 mM DTT) and centrifuged again at 5000xg for 3min to collect the cell pellet. The cell
D

pellet was re-suspended with the same lysis buffer and sonicated for 3 minutes (15 seconds of
TE

bursting with 30 seconds cooling intervals). The cell lysate was centrifuged at 14,000×g at 4°C

for 30 minutes to obtain the cell-free supernatant that was directly used in the enzyme assay. The
EP

supernatant protein was estimated by Lowry method. The ICL enzyme assay was conducted as

described elsewhere [22], with some modifications. The 1ml reaction volume was having 25 mM
C

imidazole (pH 6.8), 5 mM MgCl2, 1 mM EDTA, 4 mM phenylhydrazine HCl, 1 mM DL-


AC

isocitric acid (substrate), and cell-free extract. The reaction starts immediately after the addition

of substrate. Glyoxylate-phenylhydrazone formation was spectrophotometrically assessed at 324

nm using the VSI-501 UV-VIS spectrophotometer after incubation at 30°C to determine the ICL

activity in the reaction for the subsequent inhibition study. Similar reactions without substrate

5
ACCEPTED MANUSCRIPT

were prepared in parallel to serve as the blank. The compounds were added to the reaction at

their respective concentration followed by incubation and measurement as described above. A

drug-free control was prepared to calculate the percent inhibition of ICL enzyme activity caused

by each compound.

PT
MLS enzyme assay

RI
MLS activity was determined by the enzymatic activity assay described elsewhere [23] with

some modifications. Briefly, 1ml reaction volume was having 50 mM imidazole (pH 6.8), 100

SC
mM MgCl2, 2.5 mM Acetyl CoA, 10 mM glyoxylic acid solution, 0.2 mM 5,5'-Dithio-bis(2-

Nitrobenzoic Acid) solution (DTNB), 95% ethanol and cell-free extract. The reaction starts

U
immediately after the addition of substrate. 5-thio-2-nitrobenzoic acid (TNB) formation was
AN
spectrophotometrically assessed at 412 nm using the VSI-501 UV-VIS spectrophotometer after

incubation at 30°C to determine the MLS enzyme activity in the reaction for the subsequent
M

inhibition study. Similar reactions without substrate were prepared in parallel to serve as the
D

blank. The compounds were added to the reaction at their respective concentration followed by
TE

incubation and measurement as described above. A drug-free control was prepared to calculate

the percent inhibition of MLS enzyme activity caused by each compound.


EP

RT-PCR

RNA isolation, cDNA synthesis and reverse transcriptase RT-PCR was performed as described
C

previously [7]. The synthesized cDNA product (2µl) was directly used for PCR amplification
AC

reaction (50 µl) using gene specific forward and reverse primers (Table S1). The amplified

products were gel electrophoresed and the densities of bands (for genes of interest) were

measured and quantified by normalizing to that of the constitutively expressed actin gene

(ACT1).

6
ACCEPTED MANUSCRIPT

Maintenance and survival of Caenorhabditis elegans

Nematodes were handled according to standard method [24]. Worms were grown on NGM plate

at 20°C, unless otherwise indicated, and routinely maintained on E. coli OP50. C. elegans were

routinely purified by the bleaching method. Approximately, 40 worms at L4 stage or young adult

PT
hermaphrodites were transferred from a lawn of E.coli OP50 to BHI media in presence of PA

RI
(350 and 700 µg/mL). Worms were considered dead when they did not respond to the tapping on

the plate and was scored on daily basis for 7 days. Each experimental condition was tested in

SC
triplicate. Nematode survival was plotted with the Kaplan–Meier method.

Antifungal assay with Caenorhabditis elegans

U
For the C. elegans co-infection liquid assay, methodology with a few minor modifications was
AN
used as described elsewhere [24]. Briefly, synchronized, young adult nematodes were

sequentially preinfected with C. albicans (SC5314/Act1p-GFP) for 2 h on BHI agar medium


M

containing ampicillin (100µg/ml). The nematodes were then washed four times with 2 ml of
D

sterile M9 buffer. They were collected by centrifugation at 900 rpm between each wash and
TE

pipetted into wells of a 12-well microtiter dish (NEST Pvt Ltd) containing 2 ml liquid medium

(20% BHI and 80% M9 buffer) in presence of PA (175, 350, 700µg/ml). The infected C. elegans
EP

were incubated at 25°C for 7 days, animals were scored as live or dead on a daily basis and

images were taken on the 7th day of infection. Image was taken from microscope (Olympus) at
C

10X with equipped with Coslab camera.


AC

Visualization of Candida cells within the nematode intestine.

C. albicans, SC5314 and Act1p-GFP, lawns were prepared and worms were placed for 6 h,

thoroughly washed with M9 buffer, and then pipette onto OP50 seeded NGM plates. After 2

days worms were thoroughly washed again with M9 buffer. SC5314 fed C. elegans were stained

7
ACCEPTED MANUSCRIPT

with CFW on 2nd day of infection. Fluorescence of Act1p-GFP and CFW stained C. albicans

were observed in the proximal and distal intestine of C. elegans through HL-23 Coslab

Fluorescence microscope, India.

PT
Results and Discussion

RI
PA phenocopied ICL1 deletion mutant under low carbon utilizing conditions

SC
To check the effect of PA on GC, drop dilution assay were performed on C. albicans grown on

different non-fermentable carbon sources viz. glycerol, EtOH, citrate and acetate. Expectedly,

U
ICL1 knockout mutant (∆icl1) was hypersensitive to all the tested low carbons conditions such as
AN
acetate, citrate and ethanol and the growth was rescued similar to wild type in the revertant of

ICL1 (∆icl1+ICL1) (Fig. 1). Interestingly, C. albicans when grown in similar low carbon
M

utilizing conditions in presence of PA, it displayed hypersensitivity similar to ∆icl1 (Fig. 1).
D

These observations points out the fact that PA leads to interruption in functioning of GC. In
TE

response to glucose depletion, a major reprogramming of metabolic flux takes place.

Importantly, in our previous study some important genes, such as JEN2, MAE1 and ADH family
EP

genes that are required under low glucose condition were found to be differentially regulated in

response to PA [7]. Similarly, in our previous study, in total 6 HGT genes were found to be
C

differentially regulated, among which 5 genes (HGT4, HGT5, HGT10, HGT13, HGT18) were
AC

downregulated and 1 gene (HGT12) was upregulated [7]. Infact, HGT10 and HGT13, were

known to regulate glycerol uptake [25] and could be the reason for observed glycerol sensitivity.

The upregulated HGT12 is known to express specifically during macrophage infection and

transportation of glucose, fructose and mannose [26]. These alterations in gene expression may

provide an explanation for the observed growth defects in presence of PA on alternative carbon

8
ACCEPTED MANUSCRIPT

sources. Thus our results suggest the interruption of GC in presence of PA and that functional

GC is indispensible for C. albicans to sustain PA stress.

Three dimensional structure prediction and validation

PT
ICL and MLS models of C. albicans were predicted through I-TASSER and were obtained in

RI
PDB format. I-TASSER server is tool for protein structure and function predictions. The

predicted secondary structures were obtained with confidence core (C-score), in a range of -5 to

SC
2, a parameter to check the quality of predicted models. The higher C-score value denoted the

model with better quality (Yang and Zhang 2015). The model with C-score 1.81, TM-score 0.97

U
± 0.05, and root mean square deviation (RMSD) 3.7 ± 2.6 Å was selected as best predicted
AN
model for ICL protein (Fig. 2Ai). Similarly, the model with C-score 1.32, TM-score 0.90 ± 0.06,

and RMSD 4.7 ± 3.1 Å was selected as best model for MLS protein (Fig. 2Aii). Threading
M

templates for query proteins were identified through LOMET meta-server and estimated by
D

normalized Z-score (Table S2 and S3). Generally, if Z-score is >1 it reflects a confident
TE

alignment. However, for small sequences it usually fails to show the significant indication of

modeling accuracy. Thus, the percentage sequence identity in the threading aligned region
EP

(Iden1) and in the whole chain (Iden2) considered for the good homology (Table S2). The

structural alignment program, TM-align, identified 1dquA and 3cuzA in PDB library as best
C

structural analog for ICL and MLS with the TM-score of 0.928 and 0.946 respectively (Table
AC

S3). To check the reliability of the best predicted model, Ramachandran plot were independently

obtained from PROCHECK and MolProbity servers which indicated a good quality model for

both proteins (Fig. S1). Thus both the software justified the reliability of the predicted structure

of both the proteins.

9
ACCEPTED MANUSCRIPT

Docking studies

Auto docking 4.2 were used to determine the orientation of inhibitors bound to ICL and MLS

and the conformation with the binding energy value for each molecule was chosen for further

PT
analysis. Firstly, the active site of proteins ICL and MLS were calculated through Computed

RI
Atlas of Surface Topography of proteins (CASTp) server. CASTp evaluation observed the active

site amino acids, surface area (516.1), volume (893.5) of ICL, and the active site amino acids,

SC
surface area (1148.5) and volume (1489.6) of MLS. 96 and 95 binding pockets of ICL and MLS

were categorized to find the residues about probe 1.4Å radius (Fig. S2). Results revealed that PA

U
is binding more efficiently into the active sites of ICL and MLS with better negative binding
AN
energy (∆G) of −7.8 kcal/mol and −6.6 kcal/mol respectively as compared to known inhibitor

(Table S4). Furthermore, PA formed one H-bond interaction with Glu498 of ICL and the active
M

site residues Thr470, Ala474, Phe478 and Val 494 of ICL were involved in hydrophobic
D

interaction with PA (Figure 2Bi). However, at the active site of MLS, PA shows two H-bond
TE

interaction with Asp125, Arg174 and residues Leu256, Glu258, Gly283 and Asp286 are

involved in hydrophobic interaction (Figure 2Bii). Therefore, to finally conclude,


EP

bioinformatically it was accessed that PA have showed excellent binding energy for ICL and

MLS and it may be considered as a good inhibitor of the ICL and MLS.
C
AC

PA inhibits in vitro ICL and MLS activities and mRNA expressions

To confirm the effect of PA on the GC enzymes, ICL and MLS, the in vitro enzyme assays were

performed in the cells treated with PA. The crude extract of C. albicans cells were used to

investigate the action of PA on native ICL and MLS activities. It was noted that PA showed

10
ACCEPTED MANUSCRIPT

35.5% inhibition in the ICL enzyme activity after 35min and 59.5% inhibition in MLS enzyme

activity after 15min respectively (Fig. 3Ai and Bi). Furthermore, to determine the types of

inhibition, enzyme kinetics based on Lineweaver-Burk plot were performed in presence of PA to

check if this inhibition is competitive, non-competitive or uncompetitive. Inhibitory kinetics as

PT
revealed by Lineweaver-Burk plot displayed that PA competitively inhibits ICL activity with an

RI
increase in apparent Km from 2.08 to 4.8 with no appreciable effect on the Vmax Fig. 3Aii.

Interestingly, for MLS activity, we observed non-competitive inhibition as there was no change

SC
in Km (0.1) but Vmax was reduced from 2.6 to 1.0 (Fig. 3Bii. The inhibitory effect of PA on the

ICL1 and MLS1 genes were substantiated by performing reverse transcriptase RT-PCR. We

U
explored that the expressions of both ICL1 and MLS1 were downregulated in the cells treated
AN
with PA when compared with the untreated cells (Fig 3C). These results reinforce the hypothesis

that PA inhibits GC enzymes.


M
D

PA is non-toxic to Caenorhabditis elegans


TE

Toxicity of PA was tested by using uninfected C. elegans in vivo nematode model. It was

observed that at the MIC concentration (350µg/ml) of PA, C. elegans grew at similar pace and
EP

looks healthy as untreated control with no appreciable growth defect (Fig. 4Ai and ii). We have

further tested the toxicity of PA by hemolytic assay and observed that at its MIC concentration
C

PA showed only 10.6% hemolytic activity which depicts low toxicity (Fig. 4Aiii).
AC

PA shows antifungal activity in vivo with C. elegans infection model

We further investigated the antifungal effect of PA under in vivo condition with C. elegans

infection model. The antifungal activity of PA was tested at 1/2x MIC (175µg/ml), MIC

11
ACCEPTED MANUSCRIPT

(350µg/ml) and found that survival of C. elegans was 80% and 75%respectively, which was

enhanced when compared with survival of untreated control which was only 16% (Fig 4Bi). At

1/2x MIC concentration (175µg/ml) partial growth of C. albicans was seen but C. elegans was

surviving efficiently and were monitored as healthy which shows the loss of virulence in C.

PT
albicans (Fig 4Bii).

RI
PA reduces the persistence of C. albicans in C. elegans

SC
Persistence of C. albicans in the intestine is a critical step to express virulence during Candida

infection in C. elegans. Even the size of C. albicans is appropriate to be defecated from the

U
intestine of C. elegans [24]. Thus, we confirmed whether the MIC concentration of PA can
AN
diminish the fungal burden of C. albicans in the intestine of C. elegans. In one set of experiment

the SC5314 infected C. elegans were treated with PA and stained with calcofluor white (CFW)
M

on second day and compared with untreated worms. When observed under fluorescence
D

microscope blue colored C. albicans were clearly visible in the proximal and distal intestine of
TE

untreated C. elegans which were absent in PA treated worms (Fig. 4Ci). Furthermore, in another

set of an experiment, to substantiate this finding the constitutively expressed Act1p-GFP strain of
EP

C. albicans was fed to C. elegans and monitored for the persistence in the intestine. Presence of

C. albicans was clear from the green fluorescence both in the proximal as well as distal intestine
C

regions in untreated control which was again absent in the PA treated worms (Fig. 4Cii). This
AC

confirms that due to PA treatment C. elegans not only survives the fungal burden but can also

expel fungus out easily from its intestine to lowers its virulence activity. Together, our in vitro,

in silico and in vivo work confirms the inhibitory activity of PA on GC enzymes. In vitro study

depicts that GC enzyme activities were disrupted with the treatment of PA. In silico analyses

12
ACCEPTED MANUSCRIPT

permits an evaluation of the significant aspect to the stability and kinetics of PA to bind to the

active sites of ICL and MLS enzymes. Furthermore, in vivo studies evaluated the non toxic and

functional antifungal parameter of PA. Our results indicate that PA could be potential antifungal

candidate that warrants further attention.

PT
RI
Acknowledgment

SC
We are grateful to Joseph Heitman, Alistair Brown and Michael Lorenz for providing Candida

SC5314, constitutively expressing Act1p-GFP strain and ICL1 mutants as generous gifts

U
respectively. We thank Anindya Ghosh Roy, for providing wild-type C. elegans (N2) and
AN
Escherichia coli OP50 strains. S.H. thank for the financial assistance in the form of Young

Scientist award (SR/FT/LS-12/2012) from Science and Engineering Research Board (SERB),
M

New Delhi. Senior Research Fellowship (80/957/2015-ECD-I) from Indian Council of Medical

Research (ICMR), New Delhi to M.A.A. is deeply acknowledged. We also thank Rajendra
D

Prasad, Dean of Faculty of Science, Engineering & Technology for encouragement.


TE

Author contribution
EP

#
Both authors contributed equally to this work.
C
AC

Conflict of interest

The authors declare that they have no conflict of interest.

References

13
ACCEPTED MANUSCRIPT

1. S. Singh, Z. Fatima, S. Hameed, Predisposing factors endorsing Candida infections, Infez

Med. 23 (2015) 211-23.

2. J. Perlroth, B. Choi, B. Spellberg, Nosocomial fungal infections: epidemiology, diagnosis,

and treatment, Med Mycol. 45 (2007) 321-46.

PT
3. J. Tanwar, S. Das, Z. Fatima Z et al., Multidrug resistance: an emerging crisis, Interdiscip

RI
Perspect Infect Dis. 2014 (2014) 541340.

4. R. Prasad, K. Kapoor, Multidrug resistance in yeast Candida, Int Rev Cytol. 242 (2005) 215-

SC
48.

5. M.C. Arendrup, T.F. Patterson, Multidrug-Resistant Candida: Epidemiology, Molecular

U
Mechanisms, and Treatment, J Infect Dis. 216 (2017) S445-S51.
AN
6. M.A. Ansari, A. Anurag, Z. Fatima et al, Natural Phenolic Compounds: A Potential

Antifungal Agent, in: A. Mendez-Vilas (Eds), Book Chapter in book titled “Microbial
M

pathogens and strategies for combating them: science, technology and education”
D

(Microbiology Book Series, number # 4). Formatex Research Center, Spain.. 2013, pp. 1189-
TE

95. ISBN (13): 978-84-942134-0-3.

7. M.A. Ansari, Z. Fatima, S. Hameed, Anticandidal Effect and Mechanisms of Monoterpenoid,


EP

Perillyl Alcohol against Candida albicans, PLoS One. 11 (2016a) e0162465.

8. M.A. Ansari, Z. Fatima, S. Hameed, Cellular energy status is indispensable for perillyl
C

alcohol mediated abrogated membrane transport in Candida albicans, ADMET & DMPK. 5
AC

(2017) 126-134.

9. M.F. Dunn, J.A. Ramírez-Trujillo, I. Hernández-Lucas, Major roles of isocitrate lyase and

malate synthase in bacterial and fungal pathogenesis, Microbiology. 155 (2009) 3166-75.

14
ACCEPTED MANUSCRIPT

10. S.L. Kastora, C. Herrero-de-Dios, G.M. Avelar et al., Sfp1 and Rtg3 reciprocally modulate

carbon source-conditional stress adaptation in the pathogenic yeast Candida albicans, Mol

Microbiol. 105 (2017) 620-636.

11. L. Han, K.A. Reynolds, A novel alternate anaplerotic pathway to the glyoxylate cycle in

PT
streptomycetes, J. Bacteriol. 179 (1997) 5157–5164.

RI
12. R.S. Prado, R.J. Alves, C.M. Oliveira et al., Inhibition of Paracoccidioides lutzii Pb01

isocitrate lyase by the natural compound argentilactone and its semi-synthetic derivatives,

SC
PLoS One. 9 (2014) e94832.

13. M.A. Ansari, Z. Fatima, S. Hameed, Glyoxylate Cycle: A Promising Antimicrobial Drug

U
Target, Book Chapter in book titled “Biotechnology: Progress and Applications”, Daya
AN
Publishing House, New Delhi, 1 (2016b) 333-43. ISBN 978-93-5124-729-6.

14. M.C. Lorenz, G.R. Fink, Life and death in a macrophage: role of the glyoxylate cycle in
M

virulence, Eukaryot Cell. 1 (2002) 657-62.


D

15. M.C. Lorenz, G.R. Fink, The glyoxylate cycle is required for fungal virulence, Nature. 412
TE

(2001) 83-6.

16. M.A. Ramirez, M.C. Lorenz, Mutations in alternative carbon utilization pathways in Candida
EP

albicans attenuate virulence and confer pleiotropic phenotypes, Eukaryot. Cell. 6 (2007)

280–90.
C

17. A.M. Murad, P.R. Lee, I.D. Broadbent et al. CIp10, an efficient and convenient integrating
AC

vector for Candida albicans, Yeast. 16 (2000) 325-7.

18. S. Wu, J. Skolnick, Y. Zhang, Ab initio modeling of small proteins by iterative TASSER

simulations. BMC biology. 5 (2007) 17.

15
ACCEPTED MANUSCRIPT

19. K. Ahmad, R.A. Bhat, F. Athar, Pharmacokinetic Evaluation of Callistemon viminalis

Derived Natural Compounds as Targeted Inhibitors Against δ-Opioid Receptor and Farnesyl

Transferase, Letters in Drug Design & Discovery. 14 (2017) 488-99.

20. G.M. Morris, D.S. Goodsell, R.S. Halliday et al., Automated docking using a Lamarckian

PT
genetic algorithm and an empirical binding free energy function, J. Comput. Chem. 19

RI
(1998) 1639-62.

21. H.L. Cheah, V. Lim, D. Sandai, Inhibitors of the glyoxylate cycle enzyme ICL1 in Candida

SC
albicans for potential use as antifungal agents, PLoS One. 9 (2014) e95951.

22. S.H. Lee, T.H. Won, H. Kim et al., Suvanine sesterterpenes from a tropical sponge

U
Coscinoderma sp. inhibit isocitrate lyase in the glyoxylate cycle, Mar Drugs. 12 (2014) 5148-
AN
59.

23. R.M. Silverstein, The determination of the molar extinction coefficient of reduced DTNB,
M

Anal Biochem. 63 (1975) 281-2.


D

24. J. Breger, B.B. Fuchs, G. Aperis et al., Antifungal chemical compounds identified using a C.
TE

elegans pathogenicity assay, PLoS Pathog. 3 (2007) e18.

25. J. Horak, Regulations of sugar transporters: insights from yeast, Curr Genet. 59 (2013) 1-31.
EP

26. L. Luo, X. Tong, P.C. Farley, The Candida albicans gene HGT12 (orf19.7094) encodes a

hexose transporter, FEMS Immunol Med Microbiol. 51 (2007) 14-7.


C
AC

16
ACCEPTED MANUSCRIPT

Figure Legends

Fig. 1 Phenotypic susceptibility of PA on alternate carbon sources. C. albicans strain SC5314

(wild-type), MRC10 (∆icl1), and MRC11 (∆icl1 + ICL1) cultured on YNB agar plates

containing the indicated carbon source (2% glucose, 2% glycerol, 2% EtOH, 2% sodium citrate

PT
and 2% sodium acetate) with or without PA (175 µg/mL) for 2 days at 30 °C.

RI
Fig. 2 A) (i) Three dimensional structure of C. albicans ICL protein predicted by I-

SC
TASSER. Alignment of query protein (pink, left panel) with structural analog (cyan) 1dquA in

PDB library (right panel). (ii) Three dimensional structure of C. albicans MLS protein predicted

U
by I-TASSER (left panel). Alignment of query protein (pink) with structural analog (cyan)
AN
3cuzA in PDB library (right panel). B) Docking pose of PA (i) Surface view of PA (indicated

with black arrow) docked with ICL (left panel). Active site residues of ICL (green colored)
M

interactions with PA (right panel). (ii) Surface view of PA (indicated with red arrow) docked
D

with MLS (left panel). Active site residues of MLS (blue colored) interactions with PA (right
TE

panel). Residues are shown with ball and stick (polar interaction residue as shown in cyan and

hydrophobic interaction residue as shown in green) and compound is shown with stick model.
EP

Hydrogen bonds are shown as broken lines (black).


C

Fig 3. A. Isocitrate lyase (ICL) enzyme activity assay. i) ICL activity in presence of PA. Mean
AC

enzyme activity expressed as absorbance at 324nm + SD is depicted on Y axis. ii) Lineweaver-

Burk plot of ICL inhibition by PA. [S], substrate concentration [mM]; V, reaction velocity (∆

absorbance unit/min). Each data point represents the mean of three experiments. B. Malate

Synthase (MLS) enzyme activity assay. i) MLS activity in presence of PA. Mean enzyme

17
ACCEPTED MANUSCRIPT

activity expressed as absorbance at 412nm + SD is depicted on Y axis. ii) Lineweaver-Burk plot

of ICL inhibition by PA. [S], substrate concentration [mM]; V, reaction velocity (∆ absorbance

unit/min). Each data point represents the mean of three experiments. C. RT-PCR of ICL1 and

MLS1 in response to PA. The upper panel shows transcript levels of ICL1 and MLS1 in lanes

PT
(1) Control, (2) PA (225µg ml-1). The lower panel shows the quantitation (density expressed as

RI
Intensity/mm2) of the respective transcripts normalized with constitutively expressed ACT1

transcripts.

U SC
Fig. 4. A) Toxicity test of PA. i) Microscopic images (magnification 4x) of nematodes in
AN
presence of PA. Worm survival was determined based on movement. At least two independent

experiments were conducted. ii) Kaplan–Meier curve showing % survival of C. elegans in the
M

presence of PA. The toxicity of PA was studied on non-infected nematodes by determining

survival rates after 7 days. PA shows no toxicity on uninfected C. elegans at its MIC
D

concentration. iii) Hemolytic activity of PA depicted as percentage on Y axis in comparison to


TE

the positive control Triton X 100. B) PA prolongs the survival of C. albicans infected C.

elegans. i) Microscopic images showing the survival of infected C. elegans when treated with
EP

PA on day 3 and day 6. ii) Kaplan–Meier curve showing % survival of C. albiacns infected C.
C

elegans in the presence of PA. C) Intestinal persistence of C. albicans i) CFW stained C.


AC

albicans visualized in proximal and distal intestine of C. elegans showing lesser fungal burden

with PA treatment. ii) Act1p-GFP C. albicans visualization confirming lower fungal burden in

PA treated worms contrary to untreated nematodes.

18
ACCEPTED MANUSCRIPT

Fig. 1

PT
RI
SC5314 ∆icl1 ∆icl1+ICL1 PA

SC
Glucose

Glycerol

U
AN
EtOH
Citrate

M
Acetate

D
TE
CEP
AC
ACCEPTED MANUSCRIPT

Fig. 2
A) B)

PT
i ICL i ICL

RI
U SC
AN
M
ii MLS ii MLS

D
Gly283
Glu258

TE
EP
C
AC
ACCEPTED MANUSCRIPT

Fig. 3
A) B) C)

PT
i i
4 2.5

RI
3.5

Absorbance at 412nm
Absorbance at 324nm

2
3 Control PA

SC
2.5 1.5
2 Control Control
ACT1
1.5 1
Neg Control Neg Control
1 PA PA
ICL1

U
0.5
0.5
0 0

AN
0 5 10 15 20 25 30 35 1 5 10 15 MLS1
Time (min) Time (min)

ii ii

M
8 Control (y = 1.477x + 0.725) 4 Control
Control Control(y = 0.046x + 0.374)
7 PA (y = 3.011x + 0.626) PA 3.5 PA (y = 0.124x + 0.928) PA ACT1

D
6 3
5 2.5

1/V
ICL1
1/V

PA

TE
4 2
3 1.5 Control
2 1
0.5
MLS1
EP
1
0 0
-1 -0.5 0 0.5 1 1.5 2 2.5 -10 -5 0 5 10 15 0 0.5 1 1.5
1/[S] 1/[S]
C

Intensity/mm2
AC
ACCEPTED MANUSCRIPT

Fig. 4 A) i B) i

Control PA (MIC)

PT
ii 120 iii
ii

RI
100
Triton X 120
80
% Survival

100

SC
60

% Survival
PBS 80
40 60 Control

U
Control
20 PA 40 PA(MIC)
PA(MIC) PA(1/2 MIC)

AN
0 20
0 20 40 60 80 100 120 0
0 5 10
Time (Days) % Hemolytic activity 0 1 2 3 4 5 6 7 8

M
Time (days)

Control PA

D
C) i
CFW stained SC5314
Distal Proximal

TE
C EP
AC

ii
Proximal
Act1p-GFP

Distal
ACCEPTED MANUSCRIPT

Highlights

• PA is efficient inhibitor glyoxylate cycle of Candida albicans which is absent in humans.

• PA has negligible hemolytic activity making it viable drug candidate.

PT
• PA showed enhanced survival of C. albicans in nematode model, C. elegans demonstrating

RI
its in-vivo efficacy.

U SC
AN
M
D
TE
C EP
AC

S-ar putea să vă placă și