Computational Prediction of Leishmania Infantum Epitopes: A Bioinformatic-based Step to Leishmaniasis Vaccine Design

1. Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
2. Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
3. Department of Medical Biotechnology, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
4. Department of Oncology, Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran

Abstract

Leishmaniasis, a significant public health concern in resource-limited areas, is caused by the parasitic protozoan Leishmania. The insufficiency of current treatments underscores the urgent need for effective vaccines. Researchers have pinpointed promising vaccine targets through comprehensive antigen screening methods, showing their ability to trigger protective immune responses against Leishmania. The delicate balance between pro-inflammatory (Th1) and anti-inflammatory (Th2) responses in Leishmaniasis underscores the immune regulation complexity vital for fighting the infection. Leveraging bioinformatics tools, a multi-epitope vaccine (LIDVC) targeting KMP11, GP63, and LACK antigens aims to induce both humoral and cellular responses. In silico analysis suggests the potential of LIDVC to bolster protective immunity against Leishmania, offering a new path for vaccine development in tackling this formidable disease.

Highlights

Leishmaniasis, a significant public health concern in resource-limited areas, is caused by the parasitic protozoan Leishmania. The insufficiency of current treatments underscores the urgent need for effective vaccines. 

Full Text

Introduction
 


Leishmaniasis, a neglected tropical disease caused by the protozoan parasite Leishmania, affects millions globally, particularly in resource-limited regions (1). The intricate life cycle of Leishmania involves transmission via infected sandfly bites, resulting in diverse clinical manifestations ranging from cutaneous to visceral forms of the disease (Figure 1). The inadequacy of current treatment options underscores the urgent requirement for efficacious and safe vaccines to manage and prevent Leishmaniasis (2). Researchers have directed their efforts towards identifying antigenic proteins expressed at various stages of the parasite's life cycle to uncover novel vaccine targets through high-throughput and highly accurate methods (3, 4). This pursuit has uncovered several promising vaccine candidates, such as glycoprotein 46 (gp46), cathepsin L-like and B-like proteases, histone H2A, glucose-regulated protein 78 (grp78), and stress-inducible protein 1 (STI-1) (5). These antigens have demonstrated the ability to provoke protective immune responses against Leishmania infection in preclinical investigations. The immune response to Leishmania infection is intricate, involving a delicate equilibrium between pro-inflammatory (Th1) and anti-inflammatory (Th2) cytokines (6, 7). Th1-mediated responses, characterized by IFN-γ production, play a pivotal role in controlling parasite growth by activating macrophages to eliminate intracellular parasites (4, 7). Conversely, Th2-mediated responses can support parasite survival and disease progression. Understanding the interplay between these immune responses is vital for designing effective vaccines against Leishmaniasis. The field of bioinformatics and computational biology has transformed vaccine development by facilitating the prediction of immunodominant epitopes and antigenic proteins. In silico methodologies enable swift screening of potential vaccine candidates, reducing the time and resources needed for experimental validation (3). Leveraging these computational tools, researchers can craft epitope prediction that target specific immune pathways to bolster protective immunity against Leishmania. In a recent study, epitopes were devised for KMP11, GP63, and LACK antigens using specific bioinformatic tools.

Figure 1. Transmission and life cycle of Leishmania infantum in the vector and host body. (Design by Authors, 2024)

 

Protocol


2-1- Obtain the amino acid sequence for the target proteins

    The FASTA-formatted amino acid sequences for L. infantum GP63 (Accession no. QJF54184), KMP11 (Accession no. AGV77135), and LACK (Accession no. UQI50440) were collected from the National Center for Biotechnology Information (NCBI) website located at http://www.ncbi.nlm.nih.gov (8).

2-2- Forecasting the location of antigenic epitopes

    In this step we opted to predict CTL epitopes, linear B cell epitopes, and MHC class I and II epitopes. To do this, different bioinformatics servers capable of identifying these sorts of epitopes were used.

    a. Prediction and screening of linear B cell epitope: 

    Linear B cell epitopes were predicted utilizing the (B cell epitope prediction) section of the IEDB server which located at http://tools.iedb.org/bcell/ . The mentioned server method was also set to Bepipred Linear Epitope Prediction 2.0. BepiPred is a predictive method that identifies the location of linear B-cell epitopes. It achieves this by utilizing a hidden Markov model and propensity scale method, which allow it to determine which residues are part of an epitope based on their scores. Residues with scores higher than the threshold value of 0.35 are colored yellow on the graph and labeled with "E" in the output table. The accuracy of the prediction method for epitope/non-epitope predictions is determined by a table that summarizes data from a large benchmark calculation that included almost 85 B cell epitopes (9).

    a. Prediction and screening of linear B cell epitope: 

    Linear B cell epitopes were predicted utilizing the (B cell epitope prediction) section of the IEDB server which located at http://tools.iedb.org/bcell/ . The mentioned server method was also set to Bepipred Linear Epitope Prediction 2.0. BepiPred is a predictive method that identifies the location of linear B-cell epitopes. It achieves this by utilizing a hidden Markov model and propensity scale method, which allow it to determine which residues are part of an epitope based on their scores. Residues with scores higher than the threshold value of 0.35 are colored yellow on the graph and labeled with "E" in the output table. The accuracy of the prediction method for epitope/non-epitope predictions is determined by a table that summarizes data from a large benchmark calculation that included almost 85 B cell epitopes (9).

    b. Cytotoxic T lymphocyte (CTL) epitope prediction and screening: 

    In this section we used CTLPRED server to predict the CTL epitopes which available at http://crdd.osdd.net/raghava/ctlpred/ . The approach of this server based on two form of machine learning techniques such as Artificial Neural network (ANN) and support vector machine (SVM). Based on these two processes of server, the methods also allow for consensus and combined prediction (10).

    c. Prediction and selection approach for MHC class 1 and MHC class 2 epitopes: 

    To predict MHC class 1 epitopes, we utilized the T cell class I tool provided on the IEDB website at https://nextgen-tools.iedb.org/. The tool employs Artificial Neural Networks (ANNs) and utilizes data on 177 MHC molecules from various species such as humans, mice, cattle, primates, pigs, horses, and dogs. The IEDB recommends using percentile rank as the primary metric for ranking binding predictions, with a percentile rank of less than or equal to 1% covering 80% of the immune response for many alleles (11). In the following to anticipate MHC class 2 epitopes, the MHC2PRED at http://crdd.osdd.net/raghava/mhc2pred/ address was used. In the algorithm of this server, Support Vector Machine (SVM), a machine learning approach, was used to construct a prediction strategy for MHC binding. Individual amino acid sequences represented by binary input were used to train SVM. Each amino acid in a 9-mer peptide was turned into a 20-dimensional vector, giving each peptide a 180-dimensional vector. Non-binders were classified as -1, whereas binders were designated as +1. Experimentation was used to identify the best kernel type for data categorization, such as RBF, Polynomial, Linear, and Sigmoid. Finally, by methodically modifying the parameters and analyzing prediction performance, the kernel features and regulatory parameter C were then tuned (12).

 

Results and Discussion

 

3-1- Predicted epitopes

    a. B-Cell Epitope Prediction:

    The high-scored linear and conformational B-cell epitopes that were predicted within the full-length of the designed vaccine by the IEDB, also in combination with BepiPred linear epitope prediction 2.0, respectively (Table 1).

Table 1. B-cell epitopes predicted by the IEDB

Antigen Start End Peptide Length


KMP11
7 7 E 1
9 10 SA 2
13 14 KR 2
16 76 DEEFNRKMQALNAKFFADKPDESTLSPEMKE
HYEKFERMIKEHTEKFNKKMHEHSEHFKQK
61
78 89 AELLEQQKAAQY 12

LACK
29 48 NPDRHSVDSDYGLPSHRLEG 20
81 90 NGQCQRKFLK 10
125 135 CMHEFLRDGHE 11
168 178 GGKCERTLKGH 11


GP63
16 20 QLHTE 5
23 56 KVRQVQDKWNATGMVDEICGDFKVPPAHITEGFS 34
83 86 FSDG 4
100 106 IASRYDQ 7
126 144 FFEGARILESISNVRHKDF 19
172 191 IEDQGGAGSAGSHIKMRNAQ 20
217 219 FYQ 3

   

    b. CTL Epitopes Prediction

    The high-ranked CTL epitopes (9-mer length) with a binding affinity score were selected as final CTL epitopes in three antigens (Table 2).

Table 2. Cytotoxic T-lymphocyte (CTL) epitopes of selected antigens prediction using CTLpred server (Combined approach; Cutoff Score=0.51).

Antigen Position Sequence Score (ANN/SVM)

KMP11
7 FFADKPDES 0.990
9 KFFADKPDE 0.980
13 RLDEEFNRK 0.950

Lack
16 SHRLEGHTG 1.000
78 KFLKHTKDV 1.000
29 FVSCVSLAH 0.990

GP63
81 RILESISNV 1.000
125 PQALQLHTE 0.990
168 VRQVQDKWN 0.990
 

    c. MHC Peptide Prediction

    To predict the binding epitopes for Major Histocompatibility Complex (MHC) class I, we utilized the next-generation tools of Immune Epitope Database (IEDB, available at https://nextgen-tools.iedb.org/tc1) and focused on 9-mer length peptides and human HLAs. Specifically, based on IEDB recommended method 2020.09 (NetMHCpan EL 4.1) (13), most reference HLA allele set used for prediction; e.g. 16 class A alleles (01:01, 02:01, 02:03, 02:06, 03:01, 11:01, 23:01, 24:02, 26:01, 30:01, 30:02, 31:01, 32:01, 33:01, 68:01 and 68:02) and 11 class B alleles (07:02, 08:01, 15:01, 35:01, 40:01,44:02, 44:03, 51:01, 53:01, 57 l:01 and 58:01) (Table 3-5).

Table 3. B-cell epitopes predicted by the IEDB

Peptide Length Start End Allele Peptide index Core Icore Score
KMHEHSEHF 9 65 73 HLA-B*15:01 1577 KMHEHSEHF KMHEHSEHF 0.960679
HFKQKFAEL 9 72 80 HLA-B*08:01 1500 HFKQKFAEL HFKQKFAEL 0.955418
LEQQKAAQY 9 81 89 HLA-B*44:03 1929 LEQQKAAQY LEQQKAAQY 0.907147
LEQQKAAQY 9 81 89 HLA-B*44:02 1845 LEQQKAAQY LEQQKAAQY 0.901512
MIKEHTEKF 9 54 62 HLA-B*15:01 1566 MIKEHTEKF MIKEHTEKF 0.889601
RMIKEHTEK 9 53 61 HLA-A*03:01 389 RMIKEHTEK RMIKEHTEK 0.864294
EFNRKMQAL 9 18 26 HLA-B*08:01 1446 EFNRKMQAL EFNRKMQAL 0.828076

 

Table 4. B-cell epitopes predicted by the IEDB

Peptide Length Start End Allele Peptide index Core Icore Score
HPIVVSGSW 9 149 157 HLA-B*53:01 5093 HPIVVSGSW HPIVVSGSW 0.942054
RTLKGHSNY 9 173 181 HLA-A*30:02 2233 RTLKGHSNY RTLKGHSNY 0.91673
TLKGHSNYV 9 174 182 HLA-A*02:03 586 TLKGHSNYV TLKGHSNYV 0.913724
RTLKGHSNY 9 173 181 HLA-B*57:01 5323 RTLKGHSNY RTLKGHSNY 0.856624
KVWNVNGGK 9 162 170 HLA-A*03:01 986 KVWNVNGGK KVWNVNGGK 0.83114

 

Table 5. B-cell epitopes predicted by the IEDB

Peptide Length Start End Allele Peptide index Core Icore Score
KVRQVQDKW 9 23 31 HLA-B*57:01 5398 KVRQVQDKW KVRQVQDKW 0.991967
YLIPQALQL 9 9 17 HLA-A*02:01 224 YLIPQALQL YLIPQALQL 0.989888
YLIPQALQL 9 9 17 HLA-A*02:03 439 YLIPQALQL YLIPQALQL 0.967609
YLIPQALQL 9 9 17 HLA-A*02:06 654 YLIPQALQL YLIPQALQL 0.966576
KVRQVQDKW 9 23 31 HLA-B*58:01 5613 KVRQVQDKW KVRQVQDKW 0.963771
RILESISNV 9 131 139 HLA-A*02:06 776 RILESISNV RILESISNV 0.956299
HEMAHALGF 9 114 122 HLA-B*44:03 4844 HEMAHALGF HEMAHALGF 0.938416
RILESISNV 9 131 139 HLA-A*02:01 346 RILESISNV RILESISNV 0.923092
FSNTDFVMY 9 55 63 HLA-A*01:01 55 FSNTDFVMY FSNTDFVMY 0.911191
HEMAHALGF 9 114 122 HLA-B*44:02 4629 HEMAHALGF HEMAHALGF 0.910567

 

For MHC class II binding epitopes, we employed the MHC2PRED server and selected several peptides for each antigen, ensuring that they had a percentile rank of ≤1 and an IC50 value of ≤50. These stringent criteria were used to identify peptides with high binding scores for MHC class I and II. The specific peptides and their corresponding binding scores can be found in Table 6-8

Table 6. B-cell epitopes predicted by the IEDB

Allele Sequence Residue No. Peptide Score


HLA-DR9
RTEINLEIS 24 1.245
KFERMIKEH 95 1.185
EPRTEINLE 22 1.182
FNRKMQALN 64 1.176


HLA-DR3
EEFSAPFKR 51 1.888
RLDEEFNRK 59 1.546
HEHSEHFKQ 112 1.325
TEKFNKKMH 104 1.26


HLA-DQ7
GVKINETPL 2 1.439
EMRANEPRT 17 1.413
EFSAPFKRL 52 1.375
NIAINFANT 36 1.084


HLA-DQ8
RKMQALNAK 66 2.077
EPRTEINLE 22 1.736
ISHMANIAI 31 1.675
KQKFAELLE 119 1.618


HLA-DRB1*0901
PRTEINLEI 23 1.452
STLSPEMKE 83 1.432
HEHSEHFKQ 112 1.415
DESTLSPEM 81 1.353


HLA-DRB1*0401
FANTMMATT 41 1.556
GVKINETPL 2 1.136
YEKFERMIK 93 1.099
FFADKPDES 75 1.024

 

Table 7. B-cell epitopes predicted by the IEDB

MHC-II Alleles Predicted Epitopes
IAb VYDLESKAV, VTSLACPQQ, ATDYALTAS, GAALLWDLS, HKDNLIRVW
IAd TAISWKANP, VATERSLSV, WVTSLACPQ, DRLIVSAGR, FSPDDRLIV
IAs FVSCVSLAH, DGNTLYSGH, ICFSPSLEH, ATDYALTAS, RGWVTSLAC
IEd RVWNVAGEC, RGWVTSLAC, FSPNRFWMC, RLIVSAGRD, VSCVSLAHA
MHC-II Alleles Predicted Epitopes

 

Table 8. B-cell epitopes predicted by the IEDB

Allele Sequence Residue No. Peptide Score


HLA-DR9
KDFDVPVIN 178 1.297
KRDILVKYL 38 1.272
SVPSEEGVL 102 1.261
VDEICGDFK 73 1.246


HLA-DRB1*0101
NIAINFANT 27 2.215
VINIPAANI 128 1.671
AINFANTMK 29 1.52
LIPQALQLH 46 1.507


HLA-DQ7
SSTAVAKAR 187 1.632
IPQALQLHT 47 1.415
IPAANIASR 131 1.213
PVINSSTAV 183 1.163


HLA-DQ8
CDTLEYLEI 200 1.921
MKKRDILVK 36 1.864
ISHMANIAI 22 1.675
KKRDILVKY 37 1.574


HLA-DR13
VINIPAANI 128 1.468
HPAVGVINI 123 1.271
VGFFEGARI 160 1.269
VGVINIPAA 126 1.267


I-Ag7
MAPAAAAGY 231 1.903
ELMAPAAAA 229 1.857
NIPAANIAS 130 1.814
GFFEGARIL 161 1.785

 

    In summary, this process involved predicting and selecting potential binding epitopes for both MHC class I and II, using different servers and criteria to identify peptides with strong binding affinity to human HLAs.

    Leishmaniasis is widespread in subtropical regions, causing a significant burden annually (14). In spite of various chemotherapy and drug therapy against leishmaniasis which have some side effects, recent advances in development of efficacious vaccines seems to be an appropriate outstanding preventive strategy for improvement of the public health and infectious diseases control  (15-17).

    Epitope-prediction is the first and most basic step in the design of multi-epitopic vaccines. Today, with the advancement of new methods in epitope mapping, computer tools have increased the accuracy and speed of this process.

    In order to make these predictions as accurate as possible, after determining the immunogenic proteins in Leishmania infantum, all three prediction modes of linear B-cell epitopes, CTL epitopes and epitopes of MHC class 1 and 2 should be determined.
 

Acknowledgment


The authors are grateful to the Smart University of Medical Sciences and the technical team of JoVm for their support and we would like to acknowledge BioinfCamp.com for providing basic training along the way for this article.

 

Ethical Consideration


Not applicable.
 

Conflicts of Interest


The authors declared no conflict of interest.

 

Funding


This research received no specific grant from any funding agency in the public.

References

1. Alvar J, Vélez ID, Bern C, Herrero M, Desjeux P, Cano J, et al. Leishmaniasis Control Team. Leishmaniasis worldwide and global estimates of its incidence. PloS One. 2012;7(5):e35671. [DOI:10.1371/journal.pone.0035671] [PMID] [PMCID]
2. Pradhan S, Schwartz RA, Patil A, Grabbe S, Goldust M. Treatment options for leishmaniasis. Clin Exp Dermatol. 2022;47(3):516-21. [DOI:10.1111/ced.14919] [PMID]
3. Margaroni M, Agallou M, Tsanaktsidou E, Kammona O, Kiparissides C, Karagouni E. Immunoinformatics approach to design a multi-epitope nanovaccine against Leishmania parasite: elicitation of cellular immune responses. Vaccines. 2023;11(2):304. [DOI:10.3390/vaccines11020304] [PMID] [PMCID]
4. Yadagiri G, Singh A, Arora K, Mudavath SL. Immunotherapy and immunochemotherapy in combating visceral leishmaniasis. Front Med. 2023;10:1096458. [DOI:10.3389/fmed.2023.1096458] [PMID] [PMCID]
5. Nagill R, Kaur S. Vaccine candidates for leishmaniasis: a review. Int Immunopharmacol. 2011;11(10):1464-88. [DOI:10.1016/j.intimp.2011.05.008] [PMID]
6. Al-Qahtani AA, Alhamlan FS, Al-Qahtani AA. Pro-Inflammatory and Anti-Inflammatory Interleukins in Infectious Diseases: A Comprehensive Review. Trop Med Infect Dis. 2024;9(1):13. [DOI:10.3390/tropicalmed9010013] [PMID] [PMCID]
7. Costa-da-Silva AC, Nascimento DD, Ferreira JR, Guimarães-Pinto K, Freire-de-Lima L, Morrot A, et al. Immune responses in leishmaniasis: An overview. Trop Med Infect Dis. 2022;7(4):54. [DOI:10.3390/tropicalmed7040054] [PMID] [PMCID]
8. Sayers EW, Beck J, Bolton EE, Bourexis D, Brister JR, Canese K, et al. Database resources of the national center for biotechnology information. Nucleic Acids Res. 2021;49(D1):D10. [DOI:10.1093/nar/gkaa892] [PMID] [PMCID]
9. Jespersen MC, Peters B, Nielsen M, Marcatili P. BepiPred-2.0: improving sequence-based B-cell epitope prediction using conformational epitopes. Nucleic Acids Res. 2017;45(W1):W24-9. [DOI:10.1093/nar/gkx346] [PMID] [PMCID]
10. Bhasin M, Raghava GP. Prediction of CTL epitopes using QM, SVM and ANN techniques. Vaccine. 2004;22(23-24):3195-204. [DOI:10.1016/j.vaccine.2004.02.005] [PMID]
11. Reynisson B, Alvarez B, Paul S, Peters B, Nielsen M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 2020;48(W1):W449-54. [DOI:10.1093/nar/gkaa379] [PMID] [PMCID]
12. Lata S, Bhasin M, Raghava GP. Application of Machine Learning Techniques in Predicting MHC Binders. In: Flower, D.R. (eds) Immunoinformatics. Methods in Molecular Biology™, vol 409. Humana Press. 2007. pp. 201-15. [DOI:10.1007/978-1-60327-118-9_14] [PMID]
13. Vita R, Mahajan S, Overton JA, Dhanda SK, Martini S, Cantrell JR, et al. The immune epitope database (IEDB): 2018 update. Nucleic Acids Res. 2019;47(D1):D339-43. [DOI:10.1093/nar/gky1006] [PMID] [PMCID]
14. Knight CA, Harris DR, Alshammari SO, Gugssa A, Young T, Lee CM. Leishmaniasis: Recent epidemiological studies in the Middle East. Front Microbiol. 2023;13:1052478. [DOI:10.3389/fmicb.2022.1052478] [PMID] [PMCID]
15. Ghorbani M, Farhoudi R. Leishmaniasis in humans: drug or vaccine therapy?. Drug Des Dev Ther. 2017;12:25-40. [DOI:10.2147/DDDT.S146521] [PMID] [PMCID]
16. Ponte-Sucre A, Gamarro F, Dujardin JC, Barrett MP, López-Vélez R, García-Hernández R, et al. Drug resistance and treatment failure in leishmaniasis: A 21st century challenge. PLOS Negl Trop Dis. 2017;11(12):e0006052. [DOI:10.1371/journal.pntd.0006052] [PMID] [PMCID]
17. Srivastava S, Shankar P, Mishra J, Singh S. Possibilities and challenges for developing a successful vaccine for leishmaniasis. Parasites Vectors. 2016;9:277. [DOI:10.1186/s13071-016-1553-y] [PMID] [PMCID]