+ All Categories
Home > Documents > Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant...

Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant...

Date post: 21-Dec-2016
Category:
Upload: poonam
View: 214 times
Download: 0 times
Share this document with a friend
14
ORIGINAL PAPER Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani Arpita Kulshrestha & Vanila Sharma & Ruchi Singh & Poonam Salotra Received: 17 October 2013 /Accepted: 3 January 2014 /Published online: 22 January 2014 # Springer-Verlag Berlin Heidelberg 2014 Abstract Leishmania donovani is the causative agent of anthroponotic visceral leishmaniasis in the Indian subconti- nent. Oral miltefosine therapy has recently replaced antimo- nials in endemic areas. However, the drug is at risk of emer- gence of resistance due to unrestricted use, and, already, there are indications towards decline in treatment efficacy. Hence, understanding the mechanism of miltefosine resistance in the parasite is crucial. We employed genomic microarray analysis to compare the gene expression patterns of miltefosine- resistant and miltefosine-sensitive L. donovani . Three hundred eleven genes, representing 3.9 % of the total Leishmania genome, belonging to various functional categories including metabolic pathways, transporters, and cellular components, were differentially expressed in miltefosine-resistant parasite. Results in the present study highlighted the probable mecha- nisms by which the parasite sustains miltefosine pressure including (1) compromised DNA replication/repair mecha- nism, (2) reduced protein synthesis and degradation, (3) al- tered energy utilization via increased lipid degradation, (4) increased ABC 1-mediated drug efflux, and (5) increased antioxidant defense mechanism via elevated trypanothione metabolism. The study provided the comprehensive insight into the underlying mechanism of miltefosine resistance in L. donovani that may be useful to design strategies to increase lifespan of this important oral antileishmanial drug. Introduction Leishmania donovani is the etiological agent of visceral leish- maniasis (VL), a potentially fatal systemic protozoal infection. The emergence and spread of resistance to therapy for VL is of significance particularly in India where more than 60 % of patients do not respond to the traditional antimonial therapy. Miltefosine (hexadecylphosphocholine) is an oral drug initially developed as an anticancer agent for the treatment of cutaneous lymphomas and breast cancer that shows selec- tive activity against Leishmania (Clive et al. 1999; Croft et al. 2006). In India, miltefosine has recently taken over as the first- line therapy for VL even in areas where antimonials are effective (WHO TDR News 2004). However, widespread use of miltefosine monotherapy might lead to the rapid emer- gence of resistance in India, where VL is anthroponotic (Bryceson 2001). The long half-life of the drug may poten- tially increase the risk of development of experimental resis- tance to this drug shown to be readily induced in vitro (Seifert et al. 2003). Concerns have been raised over rise in miltefosine treatment failure and relapses (almost double) in phase IV clinical trials in India (Sundar et al. 1998; Sundar and Murray 2005). Already, reports of clinical failure and relapse have come up in India and Nepal (Sundar et al. 2006; Pandey et al. 2009). In this situation, it becomes important to under- stand the mechanism of action and development of resistance towards miltefosine in the parasite. The mechanisms and related biological pathways that con- tribute to miltefosine resistance in the parasite are relatively poorly understood. Suggested targets of miltefosine in Leishmania include perturbation of ether-lipid metabolism, glycosyl phosphatidylinositol anchor biosynthesis, signal Electronic supplementary material The online version of this article (doi:10.1007/s00436-014-3755-6) contains supplementary material, which is available to authorized users. A. Kulshrestha : V. Sharma : R. Singh : P. Salotra (*) National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi 110029, India e-mail: [email protected] Present Address: A. Kulshrestha Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA Parasitol Res (2014) 113:11711184 DOI 10.1007/s00436-014-3755-6
Transcript
Page 1: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

ORIGINAL PAPER

Comparative transcript expression analysis of miltefosine-sensitiveand miltefosine-resistant Leishmania donovani

Arpita Kulshrestha & Vanila Sharma & Ruchi Singh &

Poonam Salotra

Received: 17 October 2013 /Accepted: 3 January 2014 /Published online: 22 January 2014# Springer-Verlag Berlin Heidelberg 2014

Abstract Leishmania donovani is the causative agent ofanthroponotic visceral leishmaniasis in the Indian subconti-nent. Oral miltefosine therapy has recently replaced antimo-nials in endemic areas. However, the drug is at risk of emer-gence of resistance due to unrestricted use, and, already, thereare indications towards decline in treatment efficacy. Hence,understanding the mechanism of miltefosine resistance in theparasite is crucial. We employed genomic microarray analysisto compare the gene expression patterns of miltefosine-resistant and miltefosine-sensitive L. donovani. Three hundredeleven genes, representing ∼3.9 % of the total Leishmaniagenome, belonging to various functional categories includingmetabolic pathways, transporters, and cellular components,were differentially expressed in miltefosine-resistant parasite.Results in the present study highlighted the probable mecha-nisms by which the parasite sustains miltefosine pressureincluding (1) compromised DNA replication/repair mecha-nism, (2) reduced protein synthesis and degradation, (3) al-tered energy utilization via increased lipid degradation, (4)increased ABC 1-mediated drug efflux, and (5) increasedantioxidant defense mechanism via elevated trypanothionemetabolism. The study provided the comprehensive insightinto the underlying mechanism of miltefosine resistance in

L. donovani that may be useful to design strategies to increaselifespan of this important oral antileishmanial drug.

Introduction

Leishmania donovani is the etiological agent of visceral leish-maniasis (VL), a potentially fatal systemic protozoal infection.The emergence and spread of resistance to therapy for VL is ofsignificance particularly in India where more than 60 % ofpatients do not respond to the traditional antimonial therapy.

Miltefosine (hexadecylphosphocholine) is an oral druginitially developed as an anticancer agent for the treatmentof cutaneous lymphomas and breast cancer that shows selec-tive activity against Leishmania (Clive et al. 1999; Croft et al.2006). In India, miltefosine has recently taken over as the first-line therapy for VL even in areas where antimonials areeffective (WHO TDR News 2004). However, widespreaduse of miltefosine monotherapy might lead to the rapid emer-gence of resistance in India, where VL is anthroponotic(Bryceson 2001). The long half-life of the drug may poten-tially increase the risk of development of experimental resis-tance to this drug shown to be readily induced in vitro (Seifertet al. 2003). Concerns have been raised over rise in miltefosinetreatment failure and relapses (almost double) in phase IVclinical trials in India (Sundar et al. 1998; Sundar andMurray 2005). Already, reports of clinical failure and relapsehave come up in India and Nepal (Sundar et al. 2006; Pandeyet al. 2009). In this situation, it becomes important to under-stand the mechanism of action and development of resistancetowards miltefosine in the parasite.

The mechanisms and related biological pathways that con-tribute to miltefosine resistance in the parasite are relativelypoorly understood. Suggested targets of miltefosine inLeishmania include perturbation of ether-lipid metabolism,glycosyl phosphatidylinositol anchor biosynthesis, signal

Electronic supplementary material The online version of this article(doi:10.1007/s00436-014-3755-6) contains supplementary material,which is available to authorized users.

A. Kulshrestha :V. Sharma : R. Singh : P. Salotra (*)National Institute of Pathology (ICMR), Safdarjung HospitalCampus, New Delhi 110029, Indiae-mail: [email protected]

Present Address:A. KulshresthaRosalind Franklin University of Medicine and Science,North Chicago, IL, USA

Parasitol Res (2014) 113:1171–1184DOI 10.1007/s00436-014-3755-6

Page 2: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

transduction as well as inhibition of acyl transferase, an en-zyme involved in lipid remodeling (Lux et al. 2000). Currentevidence suggests that the drug kills Leishmania cells by aprocess reminiscent of programmed cell death (Verma andDey 2004; do Monte-Neto et al. 2011). An impairment indrug uptake machinery involving amino-phospholipidtranslocase miltefosine transporter (LdMT) and an accessoryprotein, LdRos3 (CDC50/Lem3 family) in experimentalmiltefosine-resistant Leishmania lines was proposed to bethe most likely mechanism of resistance (Perez-Victoriaet al. 2006). Proteomic analysis revealed role of eukaryoticinitiation factor eIF4 in miltefosine resistance in L. donovani(Singh et al. 2008). Regardless, a better understanding of themolecular mechanism involved is of paramount importance.

The completion of the genomic sequences of severalLeishmania species (http://www.genedb.org/) provided theopportunity to study the pattern of whole-genome differentialexpression during drug resistance. In recent times, gene ex-pression microarray has become a well-established technolo-gy by which the expression of thousands of genes can bemeasured simultaneously providing a global genetic perspec-tive on complex biological processes like drug resistance.Various studies have demonstrated the usefulness of whole-genome DNA microarrays for studying drug resistance inLeishmania (Ubeda et al. 2008; Leprohon et al. 2009; Singhet al. 2010). However, the modulations in Leishmania tran-scriptome in miltefosine resistance are poorly explored.

The current study utilized whole-genome Leishmania spp.oligonucleotide array to explore differences in gene expres-sion between miltefosine resistant and sensitive L. donovaniparasite. In this study, populations of L. donovani resistant tomiltefosine were selected in vitro in order to study global geneexpression modulation associated with resistance. In-depthbioinformatic analysis was performed to identify changes ingroups of interacting genes or pathways that may contribute toresistance to miltefosine. We have found evidence of alteredexpression of several genes belonging to DNA repair andreplication machinery, protein translation and folding, energygeneration by lipid degradation, transporter activity, and anti-oxidant defense mechanism in miltefosine-resistantL. donovani parasite.

Materials and methods

Parasite culture

Parasite isolates were prepared from bone marrow aspirate ofVL patient originating from Bihar and reporting to SafdarjungHospital (SJH), New Delhi, as described previously(Kulshrestha et al. 2011). Informed consent was obtained frompatients according to the guidelines of the Ethical Committee,SJH. This wild-type parental line (miltefosine-sensitive cell

lines referred as MIL-S1 MIL-S2 and MIL-S3) were culturedas promastigotes in Medium 199 (Sigma Aldrich, USA),25 mM HEPES N-[2-hydroxyethyl]piperazine-N−1-[2-ethanesulfonic acid] (Sigma Aldrich, USA), 100 IU, and100 μg/ml each of penicillin G (Sigma Aldrich, USA) andstreptomycin sulphate (Sigma Aldrich, USA), respectively,supplemented with 10 % heat- inactivated fetal calf serum(FCS; Gibco, USA) at 26 °C and 7.4 pH. The parasites werecharacterized as L. donovani by species-specific PCR (Salotraet al. 2001).

Preparation of miltefosine stock/drug stock

Miltefosine (Cayman Chemical Company, USA) stock wasprepared by dissolving the drug at concentration of 5 mg/ml inabsolute methanol and stored at 4 °C up to 1 month. Theworking stock was prepared fresh in Medium 199 on the dayof experiment.

Generation and characterization of experimental L. donovanistrains resistant to miltefosine

Wild-type parental L. donovani lines (MIL-S1, MIL-S2, andMIL-S3) were adapted to grow under high MIL pressure byin vitro passage with a stepwise increase in the miltefosineconcentration (2.5, 5, 7.5,10, 20, and 30 μg/ml) in mediumM199 to generate miltefosine-resistant parasite designated asMIL-R1, MIL-R2, and MIL-R3. At each step, parasites werecultured for at least five to eight passages to attain steady andoptimal cell growth comparable to its wild-type miltefosine-sensitive counterpart. TheMIL-R1 line was taken up further formicroarray analysis, while validation of microarray data byreal-time PCR was done in three miltefosine-resistant line(MIL-R1, R2, and R3). The susceptibility of miltefosine-resistant L. donovani (MIL-R1) to current antileishmanial drugs(miltefosine, SAG, amphotericin B, paromomycin, andsitamaquine) was tested at intracellular amastigote stage asdescribed previously (Singh et al. 2010; Kulshrestha et al.2011). Single-nucleotide polymorphism, in LdMT andLdRos3 genes, associated with miltefosine resistance was de-termined for both sensitive and resistant cell lines. The geneswere PCR amplified and amplicons were sequenced onAutomated Sequencer ABI 3730. Primers used for amplifica-tion and sequencing are given as ESM Tables S1.1 and S1.2.

Oligonucleotide array

One-color microarray-based gene expression profiling was car-ried out using a high-density Leishmania multispecies 60-meroligonucleotide microarray slide [8×15K format] representingthe entire genome of Leishmania infantum and Leishmaniamajor. The microarray chip, printed by Agilent Technologies,USA, included a total of 9,233 Leishmania-specific genes

1172 Parasitol Res (2014) 113:1171–1184

Page 3: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

including 540 control probes. Gene expression analysisemploying this array has been described previously (Ubedaet al. 2008; Leprohon et al. 2009; Rochette et al. 2008).

RNA isolation

Total RNA was extracted from 108 late log phasepromastigotes using Trizol reagent according to manufac-turer’s instructions. RNA clean up was performed usingRNeasy Plus mini kit (Qiagen, Gaithersburg, MD, USA) asdescribed by the manufacturer. The purified RNAwas quan-tified using Nanodrop by estimating the absorbance at 260 and280 nm. The quality and integrity of RNA was assessed onRNA 6000 Nano Assay Chips on Bioanalyzer 2100 (AgilentTechnologies, Santa Clara, CA, USA). The presence of threedistinct ribosomal peaks (18S, 24Sα, and 24Sβ) confirmedsuccessful RNA extraction.

RNA labelling and microarray hybridization

Complementary RNA (cRNA) was generated from 1 μg oftotal RNA using Quick-Amp Labeling kit (AgilentTechnologies) that directly incorporates Cy-3 labeled CTPinto the cRNA. Prehybridization and hybridization were per-formed according to manufacturers’ instructions. Labeledprobes were hybridized with Leishmania oligonucleotide ar-ray using Gene Expression Hybridization Kit (AgilentTechnologies) at 65 °C for 17 h. The hybridized arrays weresubsequently washed with gene expression hybridizationbuffers 1 and 2 using 0.005 % Triton X-102. Experimentswere performed using three independent RNA extractions.The slides were scanned immediately in Axon GenePix4000B scanner to minimize the impact of environmentaloxidants on signal intensities. GeneSpring GX 11.0.2 micro-array data and pathway analysis tool was used for data anal-ysis. Analysis involved data preprocessing; elimination ofoutliers, nonsignificantly expressed genes and false positives;and analysis of the gene lists in a biological perspective. Thedata files were in text (.txt) format and obtained fromAgilent’sFeature Extraction (FE). The summarization of the “raw”signal values was performed by computing the geometricmean. “Normalized” value was generated after log transfor-mation and normalization (scale) and baseline transformation.For each probe, the median of the log summarized values fromall the samples is calculated and subtracted from each of thesamples. Quartile (75th percentile) normalization was per-formed. Storey and bootstrapping analysis was performedfor multiple testing corrections. Statistically significant differ-entially expressed genes were determined by t test (unpaired)for two groups; (p value cutoff)<0.05. All microarray data isavailable on the GEO NCBI database in the MIAME format;http://www.ncbi.nlm.nih.gov/geo/ with the GEO accessionnumber GSE 303685. Genes with expression ratio greater

than 2.0 between wild-type and miltefosine-resistant parasitewere considered as differentially expressed were clusteredusing hierarchical clustering based on Pearson coefficientcorrelation algorithm. DNA microarray data were analyzedby custom R programs to illustrate the expression profile ofMIL-R L. donovaniby extrapolating on a chromosomemap ofL. infantum. Gene ontology annotation for functional classifi-cation of modulated genes was performed using GeneDB,BLAST2GO, and AmiGO databases. The pathway analysiswas carried out using GeneSpring GX11.0 and KEGG path-way analysis tool. String 9.05 database (string-db.org) wasemployed to understand the interaction network/patterns ofdifferentially regulated genes at protein level.

Quantitative RT-PCR

Q-RT-PCR was carried out on a selected number of genes(ESM Table S2) in order to validate the microarray experi-ments in three MIL-R lines (MIL-R1, R2, and MILR 3)generated from different parental lines to confirm the associ-ation of these genes with miltefosine resistance. Briefly, first-strand cDNA was synthesized from 5 μg of total RNA ofMIL-R/MIL-S parasites using the Superscript II RNAse Hreverse transcriptase enzyme (Invitrogen, Carlsbad, CA,USA) and Oligo dT primers (Fermentas, USA) according tothe manufacturer’s protocol. Three independent RNA prepa-rations were used for each Q-PCR experiment. Equal amountsof cDNAwere run in triplicate and amplified in 20μl reactionscontaining 1× Fast SYBR Green Mastermix (AppliedBiosystems, USA), 100 ng/ml forward and reverse primers.Reactions were carried out using ABI 7500 Real-Time PCRsystem (Applied Biosystems, USA). Initially, mixtures wereincubated at 95 °C for 10 min and then cycled 40 times at95 °C for 15 s and 60 °C for 1 min. No-template controls wereused as recommended. The relative amount of PCR productsgenerated from each primer set was determined based on thethreshold cycle (Ct) value and the amplification efficiencies.Gene expression levels were normalized to constitutivelyexpressed mRNA encoding glyceraldehyde-3-phosphate de-hydrogenase (GAPDH). Quantification of the relative changesin target gene expression was calculated using −2ΔΔCt method.Primers for targeted genes were designed using Primer ex-press software version 3.0 (Applied Biosystems, USA) andoligo-analyzer (Integrated DNATechnologies, USA).

Results

Characterization of laboratory-generated miltefosine-resistantL. donovani parasites

L. donovani field isolates were selected for miltefosine resis-tance up to 30 μg/ml drug pressure. The ED50±SEM value of

Parasitol Res (2014) 113:1171–1184 1173

Page 4: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

the MIL-S1 at promastigote stage was 7.02±0.18 μg/ml;whereas the miltefosine-resistant mutant MIL-R1 exhibited aninefold higher ED50 of 64.38±2.17 μg/ml (Table 1) com-pared to the corresponding wild-type isolate. The miltefosineresistance was stable even in the absence of drug pressure forover 12 passages. The resistance induced at promastigotestage was also evident at amastigote stage using J774.A.1murine macrophage cell line. No significant change was ob-served in the susceptibility of MIL-R parasite towards otherantileishmanial drugs (Table 1). The susceptibility ofpromastigote and amastigote forms towards miltefosine wasin good correlation (Seifert et al. 2003; Kumar et al. 2009),hence the promastigote stage of the parasite was established asan efficient model for this study.

Sequence analysis of LdMT genes revealed the previouslyreported single-nucleotide polymorphism, C1259→Aresulting in substitution of Thr 420→Asn in the MIL-R celllines. Additionally, a novel SNP, T 527→A resulting insubstitution of Val 176→Asp and in LdMT gene of resistantcell lines was observed. A nucleotide exchange leading toconversion of valine to aspartic acid is significant toperturbing protein’s structural stability. However, no pointmutations were detected in case of LdRos.

mRNA expression profiling of miltefosine-resistantL. donovani promastigote

The whole-genome Leishmaniamicroarray analysis revealed atotal of 311 differentially expressed genes representing 3.9% ofthe total Leishmaniagenome inMIL-R parasite (taking twofoldcutoff). Normalized intensity scatter plot in a base 2 logarithmicscale is shown in Fig. 1. Out of 311 genes, 184 genes (∼2.2 %)were preferentially expressed in MIL-R L. donovani, 127 genes(∼1.72 %) were found to be preferentially expressed in MIL-Sparasite. Majority of these genes showed an average change inmRNA accumulation not exceeding threefold (Table 2).

The data generated from the MIL-R Leishmania microarrayexperiments is illustrated by a chromosome map representinggene expression levels on a genomic scale (Fig. 2). A closeanalysis of the normalized expression data generated by themicroarray experiments confirmed that most genes located onthese chromosomes were modulated by a factor ranging from 2to 2.5 in mRNA abundance. The expression of genes on allchromosomes, except chromosomes 1, 2, and 3, were modulatedmore than twofold in MIL-R L. donovani, as suggested by thechromosomemap of gene expression. All modulated genes fromchromosomes 5, 15, 23, 26, and 29 were upregulated (represent-ed in red) while those of chromosome 6 were downregulated(represented in green). Interestingly, it was found that genesupregulated in miltefosine-induced resistance were mainly con-fined to chromosome numbers 15 and 23 as illustrated in Fig. 2.Table 1 Susceptibility of miltefosine resistant L. donovani mutant to-

wards antileishmanial drugs

Antileishmanial drugs ED50±SEM(μg/ml)Miltefosine-sensitiveL. donovani

ED50±SEM(μg/ml) MIL-resistantL. donovani

p value

Miltefosine Proa 7.02±0.18 64.38±2.17* 0.0003*

Amasb 3.02±0.18 >30c <0.0001*

SAG Proa 221.82±3.80 ND –

Amasb 14.65±0.67 17.76±1.38 0.15

AmphotericinB Proa 0.69±0.007 0.81±0.18 0.28

Amasb 0.44±0.01 0.52±0.14 0.08

Sitamaquine Proa 5.44±0.21 6.14±0.29 0.12

Amasb 1.09±0.13 0.91±0.15 0.17

Paromomycin Proa 14.41±0.637 19.64±1.35 0.03

Amasb 3.36±0.12 4.70±0.09 0.1

SAG sodium antimony gluconate, ND not determinedaPro Promastigotes; values represent mean of ED50±SEM of two inde-pendent experiments performed in triplicatebAmasAmastigotes; values represent mean of ED50±SEM of two inde-pendent experiments performed in triplicate at intracellular amastigotestage in J774A.1 cell linec The drug sensitivity could not be determined above this concentration asmiltefosine poses cytotoxic effect on the host macrophages above theconcentration of 30 μg/ml

*p<0.05, ED50 considered significantly different

Fig. 1 Normalized scatter plot of microarray gene expression analysisdata. The scatter plot represents normalized average signal intensityvalues (log scale) comparing MIL-R (Y-axis) versus wild-type control(X-axis). The position of each dot on the scatter plot corresponds to thenormalized average signal intensity of a single gene. Each point repre-sents an individual gene that is either upregulated in MIL resistant-relative to MIL sensitive (upper left quadrant) or that is downregulatedin MIL-resistant relative to MIL sensitive (lower right quadrant). Thecenter line represents no difference in expression level for a given genebetween the MIL sensitive and MIL resistant while the upper and lowerline indicate the 2 fold change cut off used. 3.9 % of genes were outsidethese fold-change thresholds. Linear regression analysis was performed

1174 Parasitol Res (2014) 113:1171–1184

Page 5: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

The chromosomal map representation enabled the identifi-cation of a locus on chromosome 23 that was over expressed inMIL-R parasite (Fig. 2). The upregulated genomic region com-prised of the genes hydrophilic acylated surface protein A(HASPA1 and A2) and small hydrophilic endoplasmicreticulum-associated protein (SHERP) which were all substan-tially upregulated in MIL-R parasite. SHERP proteins are re-ported to be metacyclic stage-specific and involved in mem-brane trafficking. Among the genes upregulated on chromo-some 29, Tob55, a member of topogenesis of mitochondrialouter membrane β-barrel proteins (TOB complex, also called

the sorting and assembly machinery) (Paschen et al. 2003;Gentle et al. 2004) was found upregulated in MIL-R parasitepossibly modulating translocation of ions and small moleculesacross the mitochondrial membrane in the resistant parasites.Significant genes upregulated on Chr5-included motor proteins(dynein light chain and kinesin-like protein), ATPase, andsurface antigen-like protein. The ABCA7 transporters, proteinkinase C, and tryperidoxin peroxidase were among the majorgenes preferentially expressed on chromosome 15 in MIL-Rparasite. All 17 genes upregulated on Chr26 were hypothetical.

Pathway analysis

BLAST2GO, AmiGO, and KEGG pathway analysis indicatedupregulation of DNA synthesis and transporter activities anddownregulation of protein metabolism in the MIL-R parasite.The percentage of differentially modulated genes according togene ontology (GO) function categories and variousmetabolicprocesses is shown in Fig. 3. The changes in expression levelof several genes belonging to various categories describedbelow are listed in Table 3. The interaction among the genesat protein level was analyzed using STRING 9.05 database.

DNA replication and repair mechanism

The expression of genes involved in DNA replication andrepair mechanism was found to be significantly affected.

Table 2 Patterns of Global mRNA expression in miltefosine resistantL. donovani parasite

mRNA upregulation(fold increase)

MIL-resistantL. donovani

Miltefosine-sensitiveL. donovani

2.0–2.5 90 89

2.5–3.0 47 19

3.0–3.5 23 10

3.5–4.5 16 9

4.5–6.0 8 –

Total genes 184 127

Percentage of modulated genes 2.3 % 1.6 %

The percent modulated genes calculated from the total 7,963 genesobtained in QC after filtering

Fig. 2 Chromosome wise modulation of gene expression in miltefosineresistant L. donovani. DNA microarray data were analyzed by custom Rprograms to illustrate the expression profile of L.donovaniMIL-R/WT byextrapolating on a chromosome map of L. infantum. Red lines indicateupregulated genes (greater than twofold upregulated) in MIL-R parasite,whereas green lines indicate downregulated genes (less than twofolddownregulated), gray lines are the genes exhibiting less than twofold

expression in both conditions. Chromosomes showing high upregulationinmiltefosine resistance included Chr 5, 15, and 23. The area encircledonchromosome 23 represents the locus containing genes SHERP andHASPA1. Likewise encircled locuson Chr 15 represents genes of nucleicacid synthesis and repair mechanism; on Chr 5 genes coding for cyto-skeleton molecules are encircled

Parasitol Res (2014) 113:1171–1184 1175

Page 6: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

The expression of DNA replication licensing factor(LinJ28.2490), Endo/exonuclease Mre11 (LinJ27.1550), andA/G-specific adenine glycosylase (LinJ28.2230) was down-regulated in MIL-R parasites. On the contrary, proliferativecell nuclear antigen (PCNA- LinJ15.1410) and RAD51/dmc1protein (LinJ35.5000) were overexpressed in miltefosineresistance.

Protein synthesis, folding, and secretion

MIL-S or wild-type parasite exhibited an activated proteinsynthesis in comparison toMIL-R parasite. Chaperone proteinDnaJ and ubiquitin-activating enzyme E1 involved in proteo-lytic process were downregulated in miltefosine resistance.Chaperonin TCP20 (LinJ13_V3.1400), a protein involved infolding process of proteins was also downregulated inmiltefosine resistance. Reduced expression of isoleucyl-tRNA synthetase transcript involved in aminoacyl-tRNA bio-synthesis during translation indicated downregulated tRNAbiosynthesis in MIL-R parasite. Protein-degrading enzymeswere also reduced in miltefosine resistance as suggested bydownregulated expression of proteolytic metallopeptidasesencoding genes including carboxypeptidase, metallo-peptidase, and thimet oligopeptidase. Cathepsin L-like prote-ases (LinJ08_V3.0960) were, however, upregulated in MIL-Rparasite. Among the genes involved in protein processing inER, a hypothetical protein and a putative transitional ERATPase were downregulated in miltefosine resistance.

Carbohydrate metabolism and oxidative phosphorylation

Beta-fructosidase-like protein, invertase-like protein, sucrosehydrolase-like protein (LinJ23_V3.1060) were upregulated inMIL-R parasite while 2-hydroxy-3-oxopropionate reductase,(LinJ30_V3.0170) was downregulated. Enzyme phospho-glycerate mutase (LinJ08.0060) and Acetyl CoA synthase(LinJ23.0580) were found to be upregulated in MIL-R para-site, while Pyruvate dehydrogenase E1 beta subunit(LinJ25.1790) was downregulated. Genes involved in oxida-tive phosphorylation including ATP synthase (LinJ36.3790)and vacuolar ATP synthase subunit B (LinJ28.2540) weredownregulated in MIL-R parasite.

Fatty acid biosynthesis and lipid degradation

Two genes associated with lipid metabolic process includingl ipase (L inJ31_V3.2540) and l ipase precur sor(LinJ31_V3.0870) like protein were upregulated, while fattyacid elongase (LinJ14_V3.0700), NADH-cytochrome B5 re-ductase (LinJ22_V3.0590), and 2,4-dienoyl-CoA reductase-like protein (LinJ06_V3.0960) were downregulated inmiltefosine resistance.

Transport and drug resistance

Interestingly, several genes involved in transport andtransporter-like activity were upregulated in MIL-R parasite

Fig. 3 Distribution of genes differentially modulated during miltefosineresistance in Leishmania donovani according to gene ontology (GO)function categories. a Overall distribution of GO categories of genesdifferentially expressed in MIL-R parasite suggest that genes belongingto metabolic process, transport, cell component, and organization were

affected. bDifferent metabolic processes that were modulated in MIL-Rparasite include DNA, nucleotide, nucleobase, protein and lipids.Unclassified proteins include the hypothetical proteins (proteins withunknown function and not tested experimentally) and proteins with noGO category (unclassified) that have been experimentally characterized

1176 Parasitol Res (2014) 113:1171–1184

Page 7: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

Table 3 Genes differentially modulated in miltefosine resistant Leishmania donovani parasite

S. no. Gene name Fold change[R] versus [S]

Regulation Gene description

Nucleic acid metabolic process [GO:0006259, GO:0006139, GO:0032774]

1 LinJ15_V3.1500 2.50 Up Proliferative cell nuclear antigen (PCNA), putative

2 LinJ29_V3.2710 3.22 Up Poly(A) polymerase, putative

3 LinJ23_V3.1210 3.85 Up Small hydrophilic endoplasmic reticulum-associated protein (SHERP)

4 LinJ35_V3.4950 2.09 Up RAD51/dmc1 protein (DMC1)

5 LinJ19_V3.0470 2.58 Up Nuclear cap binding complex subunit CBP30

6 LinJ21_V3.0690 2.38 Down DNA polymerase eta, putative

7 LinJ27_V3.1790 2.23 Down Endo/exonuclease Mre11, putative

8 LinJ28_V3.2550 2.93 Down DNA replication licensing factor, putative

9 LinJ11_V3.0540 2.07 Down Hypothetical protein, conserved, pseudouridine synthesis

10 LinJ17_V3.0930 2.14 Down Hypothetical protein, conserved

11 LinJ21_V3.0600 2.48 Down la RNA binding protein, putative

12 LinJ22_V3.1110 2.17 Down Ribonucleoside-diphosphate reductase small chain, putative

13 LinJ25_V3.1210 2.48 Down ATPase beta subunit, putative

14 LinJ28_V3.2290 2.56 Down A/G-specific adenine glycosylase, putative

15 LinJ28_V3.2450 2.22 Down DNA topoisomerase ii

16 LinJ32_V3.3930 2.80 Down Kinetoplast DNA-associated protein, putative

Proteinmetabolic process and catalytic activity [GO:0019538, GO:0006464, GO:0006508, GO:0003824]Protein metabolic process and catalytic activity[GO:0019538]

17 LinJ08_V3.0960 2.19 Up Cathepsin L-like protease

18 LmjF15.1480 2.57 Up cAMP specific phosphodiesterase, putative

19 LinJ23_V3.0580 2.08 Up Acetyl-CoA synthetase, putative

20 LinJ23_V3.0080 2.57 Up Agmatinase-like protein

21 LinJ20_V3.1740 2.49 Up Aminoacylase, putative, N-acyl-L-amino acid amidohydrolase, putative

22 LinJ25_V3.1540 2.80 Up Calpain family cysteine protease-like protein

23 LinJ23_V3.0700 2.16 Up Hypothetical protein

24 LmjF23.1665 2.00 Up Hypothetical protein

25 LinJ29_V3.2880 2.73 Up Hypothetical protein, conserved

26 LinJ15_V3.0050 3.53 Up Cytochrome-b5 reductase, putative

27 LinJ09_V3.1120 2.16 Up DNA-directed RNA polymerase III subunit, putative

28 LinJ29_V3.1670 2.25 Up GTPase activator protein, putative

29 LinJ19_V3.1670 2.86 Up Hypothetical protein, unknown function

30 LinJ15_V3.0170 2.80 Up Protein phosphatase 2C, putative

31 LinJ26_V3.1800 2.02 Up Hypothetical protein, conserved

32 LinJ10_V3.1360 3.22 Up Hypothetical protein

33 LinJ08_V3.0060 2.82 Up Phosphoglycerate mutase protein, putative

34 LinJ15_V3.1630 2.87 Up Protein kinase, putative

35 LinJ05_V3.1210 2.02 Up Surface antigen-like protein

36 LinJ22_V3.0630 3.08 Up Protein kinase putative, serine/threonine protein kinase sos2, putative

37 LinJ08_V3.0870 4.42 Up Protein kinase, putative

38 LinJ23_V3.0500 2.45 Up Trypanothione synthetase, putative

39 LinJ15_V3.1540 3.06 Up cAMP specific phosphodiesterase

40 LinJ15_V3.1120 2.08 Up Tryparedoxin peroxidase (TRYP)

41 LinJ12_v4.0050 2.03 Up Hypothetical protein, conserved

42 LinJ29_V3.0510 2.28 Up Hypothetical protein, unknown function

43 LinJ15_V3.0990 2.09 Up Calmodulin-like protein

44 LinJ29_V3.0910 2.07 Up Hypothetical protein, conserved

45 LinJ26_V3.0730 2.19 Up Hypothetical protein, conserved

Parasitol Res (2014) 113:1171–1184 1177

Page 8: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

Table 3 (continued)

S. no. Gene name Fold change[R] versus [S]

Regulation Gene description

46 LinJ23_V3.0590 2.13 Up Hypothetical protein

47 LinJ29_V3.1940 3.65 Up Tobb55,putative

48 LinJ16_V30930 2.20 Up Flagellar calcium-binding protein, putative

49 LinJ18_V3.0890 2.10 Up rab7 GTP binding protein, putative (Rab7)

50 LinJ26_V3.1360 2.32 Up Prefoldin-like protein

51 LinJ13_V3.1400 2.48 Down Chaperonin TCP20, putative

52 LinJ19_V3.1380 2.15 Down Phosphatidic acid phosphatase protein-like protein

53 LinJ22_V3.0590 2.53 Down NADH-cytochrome b5 reductase, putative

54 LinJ06_V3.0370 2.48 Down Glutamine synthetase, putative

55 LinJ10_V3.0660 2.17 Down Endonuclease G, putative

56 LinJ07_V3.0040 2.06 Down Hypothetical protein, conserved

57 LinJ36_V3.1420 2.23 Down Transitional endoplasmic reticulum ATPase, putative, valosin-containing protein homolog

58 LinJ06_V3.0960 2.47 Down 2,4-dienoyl-coa reductase-like protein

59 LinJ04_V3.0280 2.49 Down Hypothetical protein, conserved

60 LinJ25_V3.1040 2.80 Down Hypothetical protein, conserved

61 LinJ36_V3.0250 2.38 Down Peptidyl-prolyl cis-trans isomerase, putative

62 LinJ06_V3.1070 2.64 Down Kinesin, putative

63 LinJ34_V3.1530 2.22 Down Tyrosine phosphatase isoform, putative

64 LinJ25_V3.1790 2.38 Down Pyruvate dehydrogenase E1 beta subunit, putative

65 LinJ06_V3.0290 2.71 Down Ribonuclease H1, putative

66 LmjF22.1290 2.03 Down Ribonucleoside-diphosphate reductase small chain, putative

67 LinJ28_V3.2460 2.21 Down Hypothetical protein, conserved

68 LinJ24_V3.0460 2.48 Down Hypothetical predicted transmembrane protein

69 LinJ25_V3.1190 2.07 Down Hypothetical protein, conserved

70 LinJ21_V3.1330 2.00 Down T-complex protein 1, delta subunit, putative

71 LinJ36_V3.0230 2.44 Down SET domain protein, putative

72 LinJ28_V3.1550 2.34 Down DNA polymerase kappa, putative

73 LinJ28_V3.1180 2.40 Down Hypothetical protein, conserved

74 LinJ34_V3.3710 2.01 Down Hypothetical protein, conserved

75 LinJ32_V3.1970 2.02 Down Hypothetical protein, conserved

76 LinJ06_V3.1350 2.57 Down Hypothetical protein, unknown function

77 LinJ22_V3.0002 2.33 Down Hypothetical protein

78 LinJ36_V3.6980 2.81 Down Hypothetical protein, conserved

79 LmjF06.1290 2.30 Down Hypothetical protein, unknown function

80 LinJ18_V3.0300 2.13 Down Hypothetical protein, conserved

81 LinJ35_V3.4630 2.49 Down Hypothetical protein, conserved

82 LinJ19_V3.1380 2.15 Down Phosphatidic acid phosphatase protein-like protein

83 LinJ35_V3.0070 2.03 Down Prohibitin, putative

84 LinJ04_V3.0940 2.72 Down Chaperone protein DNAj, putative

85 LinJ13_V3.0090 4.18 Down Carboxypeptidase, putative, metallo-peptidase, Clan MA(E), family M 32

Carbohydrate metabolic process GO:0005975

86 LinJ14_V3.0180 3.25 Down Carboxypeptidase, putative, metallo-peptidase, Clan MA(E), family M32

87 LinJ23_V3.1060 2.24 Up Beta-fructosidase-like protein, invertase-like protein, sucrose hydrolase-like protein

88 LinJ30_V3.0170 2.10 Down 2-hydroxy-3-oxopropionate reductase, putative

Lipid metabolic process GO:0006629

89 LinJ31_V3.0870 3.23 Up Lipase precursor-like protein

90 LinJ31_V3.2540 2.60 Up Lipase, putative

91 LinJ14_V3.0700 2.64 Down Fatty acid elongase, putative

1178 Parasitol Res (2014) 113:1171–1184

Page 9: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

which included ABC1 (LinJ02_V3.0270) and A7(LinJ15_V3.0800) transporter. Aquaglyceroporin (AQP1)transporter (LinJ31_V3.0030) was upregulated in miltefosineresistance which is contrary to observation in antimony resis-tance where AQP1 has been reported to be downregulated(Ouellette et al. 2004). Golgi vesicular membrane trafficking(LinJ29_V3.0660) protein was also upregulated in MIL-RL. donovani. Gene coding for a surface antigen-like proteinand Rab7 GTP binding protein (LinJ18_V3.0890) involved inendocytic pathway were preferentially expressed in MIL-Rparasite. SHERP (LinJ23_V3.1210) and HASPA1, involvedin protein import in organelles and recruitment of anioniclipids were found upregulated in MIL-R parasite, whereas afew transporters including ATPase beta subunit and mitochon-drial carrier protein-like protein were downregulated in MIL-R parasite.

Signal transduction

Signal transduction and cell cycle progression molecules up-regulated in miltefosine resistance included calpain familycysteine protease-like protein (LinJ25_V3.1540), proteinphosphatase 2C (LinJ15_V3.0170), phosphatidic acidphosphatase-like protein (LinJ19_V3.1380), protein kinases[(protein kinase A and serine/threonine protein kinase sos2)LinJ22_V3.0630], and cAMP-specific phosphodiesterase(LinJ15_V3.1540).

Cytoskeleton and motor proteins

Genes coding for a dynein light chain (LinJ05_V3.0070) andkinesin-like protein (LinJ05_V3.0760) were upregulated inMIL-R parasite while a delta-tubulin gene [(d-tub)

Table 3 (continued)

S. no. Gene name Fold change[R] versus [S]

Regulation Gene description

Transporter activity [GO:0006810, GO:0005215]

92 LinJ27_V3.0940 2.53 Up ABCA7, vesicular-fusion ATPase-like protein, putative

93 LinJ06_V3.0090 2.00 Up ATP-binding cassette protein subfamily G, member 5, putative (ABCG5)

94 LinJ05_V3.0810 2.52 Up Hypothetical protein, conserved

95 LinJ36_V3.5100 2.43 Up Hypothetical protein, conserved

96 LinJ33_V3.1420 2.02 Up QA-SNARE protein putative

97 LinJ15_V3.0800 3.65 Up ATP-binding cassette protein subfamily A, member 7, putative (ABCA7)

98 LinJ29_V3.0660 2.63 Up BET1 Like protein, Golgi vesicular membrane trafficking

99 LinJ36_V3.6490 2.97 Up ADP ribosylation factor 3

100 LinJ11_V3.0550 2.15 Up Amino acid permease/transporter

101 LinJ02_V3.0270 2.90 Up ABC 1 transporter

102 LinJ05_V3.1060 2.58 Up ATPase, putative

103 LinJ29_V3.1530 2.93 Up Clathrin coat assembly protein ap19, putative, sigma adaptin, putative adaptin, putative

104 LinJ31_V3.0030 2.79 Up Aquaglyceroporin, AQP1 transporter

105 LinJ15_V3.0900 2.38 Up Nucleotide sugar transporter, putative

106 LinJ14_V3.0330 2.00 Up Hypothetical protein, amino acid transporter like activity

107 LinJ28_V3.2610 3.53 Down Vacuolar ATP synthase subunit b, putative

108 LinJ36_V3.3250 2.37 Down ATP synthase, putative

109 LinJ32_V3.1180 2.06 Down Mitochondrial carrier protein-like protein

110 LinJ32_V3.2190 2.05 Down ABC transporter-like protein

Cellular component, cell organization and biogenesis [GO:0005759, GO:0016043]

111 LinJ06_V3.0780 2.07 Up Hypothetical protein, conserved

112 LinJ25_V3.0990 2.12 Down Gamma-tubulin

113 LinJ36_V3.5220 2.12 Down Delta tubulin, putative

114 LinJ30_V3.1030 2.19 Down p22 protein precursor, putative

115 LinJ34_V3.4070 2.58 Down Hypothetical protein, conserved

Microtubule-based process GO:0007017

116 LinJ05_V3.0070 2.01 Up Dynein light chain, putative

117 LinJ05_V3.0760 2.20 Up Kinesin-like protein

Parasitol Res (2014) 113:1171–1184 1179

Page 10: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

LinJ36_V3.5220] and another kinesin protein were downreg-ulated. As described before, transcripts of calpain familycysteine protease-like protein involved in cytoskeleton remod-eling was upregulated in MIL-R parasite.

Q-PCR for validation of microarray experiments

Based on the biological significance, 14 genes were selectedfor validation by q-RT PCR. Results obtained by qRT-PCRwere consistent with the microarray data for all the genesconfirming the differential gene expression betweenmiltefosine resistant and sensitive parasites (Fig. 4).Importantly, to confirm that these modulations are associatedwith miltefosine resistance and are not due to random varia-tion, the expression of selected genes was validated in anothertwo distinct MIL-R induced L. donovani parasites (MIL-R2and MIL-R3) with respect to their corresponding wild-typeparasites, and the results in all MIL-R induced strains werefound consistent with the microarray data. Representative dataon fold changes in gene expression obtained by microarrayand Q-PCR with two MIL R cell lines is shown in Fig. 4.

Discussion

Introduction of oral drug miltefosine represents an importanttherapeutic advance in the treatment of VL in the Indiansubcontinent; however, the development of resistance remainsa serious threat. To understand the mechanism of miltefosineresistance in L. donovani, we carried out mRNA expressionprofiling using full genomic DNA microarray, a techniquesuccessfully employed to study resistance mechanisms inLeishmania (Ubeda et al. 2008; Leprohon et al. 2009; Singh

et al. 2010). Mining of the Leishmania genome revealed themodulated expression of a number of genes that might playimportant role in rendering resistance to the parasite. Weemployed promastigote stage of the parasite for the study asthere is a good correlation in the sensitivity of promastigoteand amastigote forms towards miltefosine (Seifert et al. 2003;Kumar et al. 2009). There is 99.9 % sequence similaritybetween L. infantum and L. donovani genome that justifiesthe use of this array.

The present study with miltefosine-resistant Leishmaniasuggested that the parasite adopts several strategies to counterthe toxic effect of miltefosine (summarized in Fig. 5). Themajor pathways/ mechanism affected in miltefosine resistanceincluded (1) DNA replication and repair machinery, (2) pro-tein translation process, (3) energy generation mechanism, (4)transporters, and (5) antioxidant defense mechanism. Amongthese, the antioxidant defense mechanism has recently beenreported to be operative in miltefosine resistance observed inclinical isolates (Das et al. 2013).

Altered DNA synthesis and repair mechanism

The expression of genes involved in DNA replication andrepair processes was significantly altered in miltefosine resis-tance. DNA replication licensing factor, a DNA-dependentATPase required for initiation of eukaryotic DNA replication,showed decreased expression which may lead to impairedDNA synthesis. This may be combated by enhanced expres-sion of PCNA as these proteins act as the processivity factorfor DNA polymerase delta that help tomaintain the replicationprocess uninterrupted (Lee et al. 2013). Endo/exonucleaseMre11 (Garcia et al. 2011) and A/G-specific adenineglycosylase (Denver et al. 2003) involved in DNA double-

Fig. 4 Validation of DNAmicroarrays expression data byquantitative real-time PCR (qRT-PCR). The gene expressionprofile of selected genes frommicroarray data (green bars) arecompared to qRT-PCR data intwo distinct induced miltefosine-resistant strains (MIL-R1 and 2).Fold changes in gene expressionfor two of the MIL-R parasites,MIL-R1 (red bar) and MIL-R2(blue bar) with respect to MIL-sensitive strains (MIL-S1 andMIL-S2), are represented here.The qRT-PCR data werenormalized with GAPDH gene

1180 Parasitol Res (2014) 113:1171–1184

Page 11: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

strand break repair and in base excision repair, respectively,were downregulated, suggesting that parasite adopts bypassprocess for DNA repair and synthesis. However, resistant cellsmay carry out essential DNA repair with the help of upregu-lated RAD51/dmc1 protein (Le Calvez-Kelm et al. 2012), anonsequence-specific DNA binding protein involved in DNArepair. The above facts indicated the compensatory behaviorof parasite machinery to continue the replication processuninterrupted in miltefosine resistance.

Reduced protein synthesis and degradation

Genes involved in translation, protein folding, and degrada-tion were found downregulated in MIL-R parasites. The find-ings suggested increased half-life of proteins in MIL-R para-site since protein synthesis as well as degradation was down-regulated. Downregulation of isoleucyl-tRNA synthetase tran-script indicated reduced protein synthesis, which may becompensated by low proteolytic activity indicated by reducedexpression of chaperone protein DnaJ and ubiquitin-activatingenzyme E1 involved in proteasome-mediated proteolytic pro-cess. Genes involved in protein phosphorylation pathway,signal transduction, and cell cycle progression, such as proteinphosphatase 2C, protein kinase A, and serine/threonine pro-tein kinase sos2, were upregulated during miltefosine resis-tance. Interestingly, prefoldin chaperone transcript was upreg-ulated in miltefosine resistance suggesting compensation forreduced TCP20 and DnaJ chaperons.

In protozoan parasites, cathepsin L proteases have beenknown to play an important role in the infection, replication,development, and metabolism (Mottram et al. 1998). Theelevated levels of cathepsin L transcript in the current studysuggested its role in miltefosine resistance. In cancer, doxoru-bicin resistance was prevented in the presence of inhibitor tocathepsin L, suggesting that inhibition of this enzyme reversesthe development of drug resistance (Zheng et al. 2009).Targeting this enzyme family in miltefosine resistance maytherefore be one strategy in the development of new VLchemotherapy.

Altered energy generation

We did not observe changes in the key enzymes of glycolyticpathway indicating uninterrupted production of pyruvate; how-ever, downregulated expression of Pyruvate dehydrogenase E1beta subunit, the first enzyme of pyruvate dehydrogenase com-plex (PDC) may lead to reduced production of acetyl CoA. Onthe contrary, the upregulated activity of Acetyl-CoA synthasemay promote synthesis of acetyl CoA to produce energy andelectron carriers. However, in MIL-R parasite, ATP productionmay be affected due to downregulation of ATP synthase andvacuolar ATP synthase subunit B, multisubunit proton pumps(Nishi and Forgac 2002) components of oxidative

phosphorylation pathway. Thus, miltefosine pressure eventuallymay lead to reduced energy generation by oxidative phosphor-ylation and the parasite may have to rely on an alternate sourceof energy. The upregulated expression of both lipase and lipaseprecursor-like protein coupled with downregulation of enzymesin lipid biosynthesis (fatty acid elongase and NADH-cytochrome B5 reductase) indicated that lipid catabolism maybe utilized for energy generation in miltefosine resistance.

Upregulation of transporters

Several membrane modifications were indicated inmiltefosine resistance, at the level of plasma membrane (asdepicted by upregulated expression of ABC transporters andHASPAs) as well as mitochondria (SHERP and Tobb55) andendoplasmic reticulum (SHERP; Fig. 5).

The present data showed upregulated expression of ABC1transporters (ABCA7, ABCG5, and ABCG2) in miltefosineresistance. Antimonial resistance was associated with overex-pression of ABC efflux pumps P-glycoproteins (Legare et al.2001) in Leishmania. ABCG-like transporter has a role in drugresistance and transbilayer lipid movement (Castanys-Munozet al. 2007), and its overexpression was shown to affect phos-pholipid trafficking (Castanys-Munoz et al. 2008; Vasiliou et al.2009). An inhibitor of ABC transporters may allow accumula-tion of miltefosine in the parasite and may serve as one of thepossible strategies to combat miltefosine resistance. Proteinkinases A or C have been demonstrated to phosphorylateABC transporters, including P-glycoprotein (Abe-Dohmaeet al. 2004), and ABCA1 (See et al. 2002; Martinez et al.2003; Yamauchi et al. 2003). Upregulated transcripts of proteinkinases in the present study suggested that these may possiblybe modulating the activity of ABC transporters.

SHERP, a 6.2-kDa small hydrophilic endoplasmicreticulum-associated protein, is exclusively expressed onlyin the metacyclic parasites (Brodin et al. 1992; Flinn andSmith 1992; Coulson et al. 1997). It shows weak associationwith the ER and outer mitochondrial membrane and is expect-ed to be involved in various cellular processes such as proteinimport in organelles and recruitment of anionic lipids duringmetacyclogenesis (Knuepfer et al. 2001). The upregulatedexpression of this gene in MIL-R parasite observed in thisstudy indicates the possibility of increased parasite fitness inresistant strains. Upregulated expression of Tob55 mitochon-drial outer membrane protein in the present study indicatedmodulated translocation of ions and small molecules acrossthe mitochondrial membrane in miltefosine resistant parasites.

Antioxidant defense mechanism

Generation of oxidants has been identified as the primarymechanism of drug-induced cell death. The parasite adoptsvarious defense mechanisms to cope with drug-induced

Parasitol Res (2014) 113:1171–1184 1181

Page 12: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

oxidative stress by expressing antioxidant proteins such asHSP70 (Brochu et al. 2004) and HSP83 (Vergnes et al.2007) that decrease superoxide radical production and therebythe cell damage. Upregulated expression of prefoldin chaper-one family observed here may likewise be contributing inparasites’ defence against miltefosine pressure (Martin-Benito et al. 2002). Downregulation of prohibitin, a moleculeinvolved in Leishmania–host interaction, observed in the pres-ent study indicated that the MIL-R cells may survive bysuppressing the expression of pro-apoptotic proteins (Jainet al. 2010; Welburn and Murphy 1998).

The ability of trypanosomatids to transform glutathione(GSH) into trypanothione (T[SH]2) is critical for vitality andvirulence of the parasite. T(SH)2, known to enhance anti-oxidant metabolism in Leishmania during drug resistance(Miller et al. 2000; Oza et al. 2003), was found upregulated inMIL-R parasite. Trypanothione synthetase (TrS), a rate-limitingenzyme in the conversion of GSH to T[SH]2, was upregulatedin MIL-R parasite. T[SH]2 has been reported to play a role inantimony resistance inLeishmania (Oza et al. 2003; Goyeneche-

Patino et al. 2008) and the current data suggested its role inresistance towards miltefosine. In Leishmania, T[SH]2 providesreducing equivalents to facilitate the parasite’s defence againstoxidative stress with enzymes of the tryparedoxin peroxidase(TryP) family principally responsible for detoxification of per-oxides (Fairlamb and Cerami 1992; Flohe et al. 1999).Enhanced antioxidant defences indicated by elevated levels ofTryP were observed in antimony and arsenite resistantLeishmania parasites (Lin et al. 2005; Wyllie et al. 2010).Upregulated expression of TryP, TrS, and T[SH]2 in MIL-Rparasite implied that enhanced antioxidant defences, may wellbe a key feature of resistance to miltefosine.

In conclusion, MIL-R parasite uses alternative strategiesfor uninterrupted DNA replication, protein synthesis andproper folding, meeting energy demand of the cell, transportof ions and small molecules, and antioxidant defense mecha-nism to subvert the drug effects. The study contributes to-wards understanding perturbed biochemical behavior ofLeishmania in response to high miltefosine pressure. Theinterdependent regulation of gene expression to combat the

Fig. 5 Model depicting mechanism of miltefosine resistance inL. donovani. Genes altered in MIL-R parasite are represented. Genesmarked with arrow represent the upregulated genes and the ones markedwith arrow represent the downregulated genes in case of MIL-R parasite.1, 2, 3, 4, 5, and 6 The probable mechanisms of resistance in the MIL-Risolate. 1 ABC 1 transporters (ABCA1 and A7) upregulated in MIL-Rthat lead to efflux of MIL out of the cell. Protein kinase (PK), known tophosphorylate the ABC1 transporters, stabilize the expression of theseproteins on plasma membrane, possibly contributing to MIL-R. 2Lipasesupregulated inMIL-R are involved in fatty acid metabolism and free fattyacids (FFA) from lipid degradation could be destined to beta oxidation forenergy generation as an alternate energy source. 3 Upregulation of

AcetylcoA synthetase (involved in TCA cycle) in MIL-R. 4Upregulationof trypanothione [T(SH)2] biosynthesis by trypanothione synthetase(TrS) in MIL-R enhances the anti-oxidant metabolism of Leishmaniaparasites thereby contributing to MIL resistance. Upregulation oftryparedoxin peroxidase [TP(ox)], responsible for hydroperoxide detox-ification further aids in antioxidant defense in MIL-R. 5 Altered DNAsynthesis indicated by decreases expression of DNA replication licensingfactor may be combated by increased expression of PCNA and DNArepair enzymes RAD51/dmc1. 6Reduced protein translation, folding, anddegradation indicated by reduced expression of aminoacyltRNAsynthatase, cheparon proteins, and ubiquitin activating enzymes

1182 Parasitol Res (2014) 113:1171–1184

Page 13: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

drug pressure indicates that development of parasite’s resis-tance towards a drug is multifactorial phenomenon that helpsparasite to sustain the lethal effects of miltefosine. Suchknowledge could be helpful in monitoring resistance in clin-ical isolates or to subsequently target major molecular inter-actions associated with miltefosine resistance.

Acknowledgments We are thankful to Dr Marc Ouellette at the Re-search Centre in Infectious Diseases, Faculty of Medicine, Laval Univer-sity, Quebec, Canada for kindly sharing the microarray design with us.AK and VS are grateful to Council for Scientific and Industrial Researchand University Grants Commission, India, respectively, for providingresearch fellowship. This work was supported by Indian Council ofMedical Research grant number 63/4/2007/-BMS.

Conflict of interest The authors do not have commercial or otherassociations that might pose any competing interest.

References

Abe-Dohmae S, Ikeda Y, Matsuo M, Hayashi M, Okuhira K, Ueda K,Yokoyama S (2004) ABCA7 supports apolipoprotein-mediated re-lease of cellular cholesterol and phospholipid to generate highdensity lipoprotein. J Biol Chem 279:604–611

Brochu C, Haimeur A, OuelletteM (2004) The heat shock protein HSP70and heat shock cognate protein HSC70 contribute to antimonytolerance in the protozoan parasite Leishmania. Cell StressChaperones 9:294–303

Brodin TN, Heath S, Sacks DL (1992) Genes selectively expressed in theinfectious (metacyclic) stage of Leishmania major promastigotesencode a potential basic-zipper structural motif. Mol BiochemParasitol 52:241–250

Bryceson A (2001) A policy for leishmaniasis with respect to the pre-vention and control of drug resistance. Trop Med Int Health 6:928–934

Castanys-Munoz E, Alder-Baerens N, Pomorski T, Gamarro F, CastanysS (2007) A novel ATP-binding cassette transporter from Leishmaniais involved in transport of phosphatidylcholine analogues and resis-tance to alkyl-phospholipids. Mol Microbiol 64(5):1141–1153

Castanys-Munoz E, Perez-Victoria JM, Gamarro F, Castanys S (2008)Characterization of an ABCG-like transporter from the protozoanparasite Leishmania with a role in drug resistance and transbilayerlipid movement. Antimicrob Agents Chemother 52(10):3573–3579

Clive S, Gardiner J, Leonard RC (1999)Miltefosine as a topical treatmentfor cutaneous metastases in breast carcinoma. Cancer ChemotherPharmacol 44:S29–S30

Coulson RM, Connor V, Ajioka JW (1997) Using 3′ untranslated se-quences to identify differentially expressed genes in Leishmania.Gene 196:159–164

Croft SL, Seifert K, Yardley V (2006) Current scenario of drug develop-ment for leishmaniasis. Indian J Med Res 123:399–410

Das M, Saudagar P, Sundar S, Dubey VK (2013) Miltefosine-unresponsive Leishmania donovani has a greater ability thanmiltefosine-responsive L. donovani to resist reactive oxygen species.FEBS J 280:4807–4815

Denver DR, Swenson SL, Lynch M (2003) An evolutionary analysis ofthe helix-hairpin-helix superfamily of DNA repair glycosylases.Mol Biol Evol 20:1603–1611

do Monte-Neto RL, Coelho AC, Raymond F, Legare D, Corbeil J, MeloMN, Frezard F, Ouellette M (2011) Gene expression profiling and

molecular characterization of antimony resistance in Leishmaniaamazonensis. PLoS Negl Trop Dis 5:e1167

Fairlamb AH, Cerami A (1992) Metabolism and functions oftrypanothione in the Kinetoplastida. Annu Rev Microbiol 46:695–729

Flinn HM, Smith DF (1992) Genomic organisation and expression of adifferentially-regulated gene family from Leishmania major.Nucleic Acids Res 20:755–762

Flohe L, Hecht HJ, Steinert P (1999) Glutathione and trypanothione inparasitic hydroperoxide metabolism. Free Radic Biol Med 27:966–984

Garcia V, Phelps SE, Gray S, Neale MJ (2011) Bidirectional resection ofDNA double-strand breaks by Mre11 and Exo1. Nature 479:241–244

Gentle I, Gabriel K, Beech P, Waller R, Lithgow T (2004) The Omp85family of proteins is essential for outer membrane biogenesis inmitochondria and bacteria. J Cell Biol 164:19–24

Goyeneche-Patino DA, Valderrama L, Walker J, Saravia NG (2008)Antimony resistance and trypanothione in experimentally selectedand clinical strains of Leishmania panamensis. Antimicrob AgentsChemother 52:4503–4506

Jain R, Ghoshal A, Mandal C, Shaha C (2010) Leishmania cell surfaceprohibitin: role in host-parasite interaction. Cell Microbiol 12:432–452

Knuepfer E, Stierhof YD, McKean PG, Smith DF (2001)Characterization of a differentially expressed protein that shows anunusual localization to intracellular membranes in Leishmaniamajor. Biochem J 356:335–344

Kulshrestha A, Singh R, Kumar D, Negi NS, Salotra P (2011) Antimony-resistant clinical isolates of Leishmania donovani are susceptible toparomomycin and sitamaquine. Antimicrob Agents Chemother 55:2916–2921

Kumar D, Kulshrestha A, Singh R, Salotra P (2009) In vitro susceptibilityof field isolates of Leishmania donovani to Miltefosine andamphotericin B: correlation with sodium antimony gluconate sus-ceptibility and implications for treatment in areas of endemicity.Antimicrob Agents Chemother 53:835–838

Le Calvez-Kelm F, Oliver J, Damiola F, Forey N, Robinot N, Durand G,Voegele C, Vallée MP, Byrnes G, Registry BC, Hopper JL, SoutheyMC, Andrulis IL, John EM, Tavtigian SV, Lesueur F (2012) RAD51and breast cancer susceptibility: no evidence for rare variant associ-ation in the Breast Cancer Family Registry study. PLoS One 7:e52374

Lee KY, Fu H, Aladjem MI, Myung K (2013) ATAD5 regulates thelifespan of DNA replication factories by modulating PCNA levelon the chromatin. J Cell Biol 200:31–44

Legare D, Cayer S, Singh AK, Richard D, Papadopoulou B, Ouellette M(2001) ABC proteins of Leishmania. J Bioenerg Biomembr 33:469–474

Leprohon P, Legare D, Raymond F, Madore E, Hardiman G, Corbeil J,Ouellette M (2009) Gene expression modulation is associated withgene amplification, supernumerary chromosomes and chromosomeloss in antimony-resistant Leishmania infantum. Nucleic Acids Res37:1387–1399

Lin YC, Hsu JY, Chiang SC, Lee ST (2005) Distinct overexpression ofcytosolic and mitochondrial tryparedoxin peroxidases results inpreferential detoxification of different oxidants in arsenite-resistantLeishmania amazonensiswith and without DNA amplification. MolBiochem Parasitol 142:66–75

Lux H, Heise N, Klenner T, Hart D, Opperdoes FR (2000) Ether–lipid(alkyl-phospholipid) metabolism and the mechanism of action ofether–lipid analogues in Leishmania. Mol Biochem Parasitol 111:1–14

Martin-Benito J, Boskovic J, Gomez-Puertas P, Carrascosa JL, SimonsCT, Lewis SA, Bartolini F, Cowan NJ, Valpuesta JM (2002)Structure of eukaryotic prefoldin and of its complexes with unfoldedactin and the cytosolic chaperonin CCT. EMBO J 21:6377–6386

Martinez LO, Agerholm-Larsen B, Wang N, Chen W, Tall AR (2003)Phosphorylation of a pest sequence in ABCA1 promotes calpain

Parasitol Res (2014) 113:1171–1184 1183

Page 14: Comparative transcript expression analysis of miltefosine-sensitive and miltefosine-resistant Leishmania donovani

degradation and is reversed by ApoA-I. J Biol Chem 278:37368–37374

Miller MA, McGowan SE, Gantt KR, Champion M, Novick SL,Andersen KA, Bacchi CJ, Yarlett N, Britigan BE, Wilson ME(2000) Inducible resistance to oxidant stress in the protozoanLeishmania chagasi. J Biol Chem 275:33883–33889

Mottram JC, Brooks DR, Coombs GH (1998) Roles of cysteine protein-ases of trypanosomes and Leishmania in host-parasite interactions.Curr Opin Microbiol 1:455–460

Nishi T, Forgac M (2002) The vacuolar (H+)-ATPases—nature’s mostversatile proton pumps. Nat Rev Mol Cell Biol 3:94–103

Ouellette M, Drummelsmith J, Papadopoulou B (2004) Leishmaniasis:drugs in the clinic, resistance and new developments. Drug ResistUpdat 7:257–266

Oza SL, Ariyanayagam MR, Aitcheson N, Fairlamb AH (2003)Properties of trypanothione synthetase from Trypanosoma brucei.Mol Biochem Parasitol 131:25–33

Pandey BD, Pandey K, Kaneko O, Yanagi T, Hirayama K (2009) Relapseof visceral leishmaniasis after miltefosine treatment in a Nepalesepatient. Am J Trop Med Hyg 80:580–582

Paschen SA, Waizenegger T, Stan T, Preuss M, Cyrklaff M, Hell K,Rapaport D, Neupert W (2003) Evolutionary conservation of bio-genesis of beta-barrel membrane proteins. Nature 426:862–866

Perez-Victoria FJ, Sanchez-Canete MP, Castanys S, Gamarro F (2006)Phospholipid translocation and miltefosine potency require bothL. donovanimiltefosine transporter and the new protein LdRos3 inLeishmania parasites. J Biol Chem 281:23766–23775

Rochette A, Raymond F, Ubeda JM, Smith M, Messier N, Boisvert S,Rigault P, Corbeil J, Ouellette M, Papadopoulou B (2008) Genome-wide gene expression profiling analysis of Leishmania major andLeishmania infantum developmental stages reveals substantial dif-ferences between the two species. BMC Genomics 9:255. doi:10.1186/1471-2164-9-255

Salotra P, Sreenivas G, Pogue GP, Lee N, Nakhasi HL, Ramesh V, NegiNS (2001) Development of a species-specific PCR assay for detec-tion of Leishmania donovani in clinical samples from patients withkala-azar and post-kala-azar dermal leishmaniasis. J Clin Microbiol39:849–854

See RH, Caday-Malcolm RA, Singaraja RR, Zhou S, SilverstonA, HuberMT, Moran J, James ER, Janoo R, Savill JM, Rigot V, Zhang LH,WangM, Chimini G,Wellington CL, Tafuri SR, HaydenMR (2002)Protein kinase A site-specific phosphorylation regulates ATP-binding cassette A1 (ABCA1)-mediated phospholipid efflux. JBiol Chem 277:41835–41842

Seifert K, Matu S, Javier Perez-Victoria F, Castanys S, Gamarro F, CroftSL (2003) Characterisation of Leishmania donovani promastigotesresistant to hexadecylphosphocholine (miltefosine). Int JAntimicrob Agents 22:380–387

Singh G, Chavan HD, Dey CS (2008) Proteomic analysis of miltefosine-resistant Leishmania reveals the possible involvement of eukaryoticinitiation factor 4A (eIF4A). Int J Antimicrob Agents 31:584–586

Singh R, Kumar D, Duncan RC, Nakhasi HL, Salotra P (2010)Overexpression of histone H2A modulates drug susceptibility inLeishmania parasites. Int J Antimicrob Agents 36:50–57

Sundar S,MurrayHW (2005)Availability of miltefosine for the treatmentof kala-azar in India. Bull World Health Organ 83:394–395

Sundar S, Rosenkaimer F, Makharia MK, Goyal AK, Mandal AK, VossA, Hilgard P, Murray HW (1998) Trial of oral miltefosine forvisceral leishmaniasis. Lancet 352:1821–1823

Sundar S, Kumar K, Chakravarty J, Agrawal D, Agrawal S, Chhabra A,Singh V (2006) Cure of antimony-unresponsive Indian post-kala-azar dermal leishmaniasis with oral miltefosine. Trans R Soc TropMed Hyg 100:698–700

Ubeda JM, Legare D, Raymond F, Ouameur AA, Boisvert S, Rigault P,Corbeil J, Tremblay MJ, Olivier M, Papadopoulou B, Ouellette M(2008) Modulation of gene expression in drug resistant Leishmaniais associated with gene amplification, gene deletion and chromo-some aneuploidy. Genome Biol 9:R115

Vasiliou V, Vasiliou K, Nebert DW (2009) Human ATP-binding cassette(ABC) transporter family. Hum Genomics 3(3):281–290

Vergnes B, Gourbal B, Girard I, Sundar S, Drummelsmith J, Ouellette M(2007) A proteomics screen implicates HSP83 and a smallkinetoplastid calpain-related protein in drug resistance inLeishmania donovani clinical field isolates by modulating drug-induced programmed cell death. Mol Cell Proteomics 6:88–101

Verma NK, Dey CS (2004) Possible mechanism of miltefosine-mediateddeath of Leishmania donovani. Antimicrob Agents Chemother 48:3010–3015

Welburn SC, Murphy NB (1998) Prohibitin and RACK homologues areup-regulated in trypanosomes induced to undergo apoptosis and innaturally occurring terminally differentiated forms. Cell DeathDiffer 5:615–622

World Health Organisation, Geneva, Switzerland. Countries of South-East Asia region plan to eliminate kala azar. TDR News Oct 2004

Wyllie S, Mandal G, Singh N, Sundar S, Fairlamb AH, Chatterjee M(2010) Elevated levels of tryparedoxin peroxidase in antimonyunresponsive Leishmania donovani field isolates. Mol BiochemParasitol 173:162–164

Yamauchi Y, Hayashi M, Abe-Dohmae S, Yokoyama S (2003)Apolipoprotein A-I activates protein kinase C alpha signaling tophosphorylate and stabilize ATP binding cassette transporter A1 forthe high density lipoprotein assembly. J Biol Chem 278:47890–47897

Zheng X, Chu F, Chou PM, Gallati C, Dier U, Mirkin BL, Mousa SA,Rebbaa A (2009) Cathepsin L inhibition suppresses drug resistancein vitro and in vivo: a putative mechanism. Am J Physiol CellPhysiol 296:C65–C74

1184 Parasitol Res (2014) 113:1171–1184


Recommended