year 16, Issue 5 (September - October 2022)                   Iran J Med Microbiol 2022, 16(5): 430-446 | Back to browse issues page


XML Persian Abstract Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Karimi Rouzbahani A, Kheirandish F, Hashemzadeh P. Bioinformatics Analysis to Designing a Multi-epitope-based Peptide Vaccine Combat Leishmania major. Iran J Med Microbiol 2022; 16 (5) :430-446
URL: http://ijmm.ir/article-1-1596-en.html
1- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
2- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
3- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran , pejman7genetian@gmail.com
Full-Text [PDF 1713 kb]   (689 Downloads)     |   Abstract (HTML)  (1683 Views)
Full-Text:   (1151 Views)
Introduction


Leishmaniasis is one of the most important infectious diseases, and zoonosis caused by Leishmania parasites and is the most important tropical disease after malaria (1, 2). Leishmaniasis is one of the primary transferable global health issues in many tropical and subtropical countries and it is endemic in 102 countries (3-5). According to the World Health Organization (WHO) report, more than 350 million people are at risk of leishmaniasis infection. Annually 200,000-400,000 people have visceral leishmaniasis (VL) and 700,000-1200,000 people have cutaneous leishmaniasis (CL). The disease has now spread to some non-endemic regions. The disease burden (daily) is reported to be 3.3 million people; clinical manifestations are cutaneous (CL), cutaneous mucosal (MCL), and visceral (VL) (6-10).
Cutaneous leishmaniasis (CL) is the most common form of leishmaniasis, transmitted to humans by biting a female sandfly. One of the causes of this disease is the intracellular parasite Leishmania major (11, 12). Cutaneous leishmaniasis causes ugly ulcers that remain in place for a long time. It also leaves scars after healing and, in terms of beauty and psychological effects on the patient, causes major problems (13, 14).
The only treatments available for cutaneous leishmaniasis are pentavalent antimonials, amphotericin B, and pentamidine. However, drug-resistant and drug toxicity are two serious concerns, which inevitably limit the chemotherapeutic options. Hence, preventive strategies such as vaccines may seem more useful in combating cutaneous leishmaniasis (15). Unfortunately, despite all the efforts made by applying different vaccination strategies, no vaccine is available to humans available against leishmaniasis (16-18).
Scientists have studied the production of various vaccines, such as a killed parasite, subunit, and DNA (19). A small number of vaccines available to combat Leishmania parasites have entered the clinical phase, so many studies are needed to find a way to treat leishmaniasis (13).
The most important factor in designing a strong vaccine for CL, is identifying the host immunity against L. major (20). During CL infection, the leishmaniasis parasite enters the macrophage of an infected person, activating the Th1 and Th2 responses. Th1 responses produce interferon-gamma and IL-2. Th2 responses produce antibodies and cytokines IL-10 and IL-4 (21).
One of the significant factors in immunization is that there are epitopes in vaccination that the immune system recognizes. The epitope is the fragment of the antigen which is detected by the immune system. Therefore, in the structure of novel vaccines (Multiepitope peptide vaccines), the identification of immunogenic epitopes could be very practical (22, 23).
Bioinformatics tools have boosted the recognition of the epitopes (24). Bioinformatics is modern science that uses computers, computer software, and databases to answer biological problems, especially in the cellular and molecular fields. Bioinformatics is an online tool based on various databases and algorithms, which is created and applied to predict protein structures, cellular and molecular properties, epitopes, and more (25, 26).
Identification of effective antigens and appropriate adjuvants are able to increase the potency and immunogenicity of novel vaccines (27, 28).
In this study, LACK, CPB, and KMP-11 proteins, which are candidate proteins for the CL vaccine, were selected for the study.
Kinetoplastid membrane protein 11 (KMP11) is a complex protein highly associated with Leishmania promastigotes lipophosphoglycan. Lipophos-phoglycan is strongly antigenic to human T cells. KMP11 is a protein specific to Kinetoplastida, capable of building innate and adaptive immunity against Leishmania. Among the different Leishmania molecules known as possible vaccine antigens, KMP-11 has attracted much attention due to its high human T cell antigen (29-31).
Another antigen is LACK (Leishmania homologue of receptors for Activated C Kinase), which plays a prominent role in the immunopathogenesis of Leishmania major by generating a Th2 response. T cells produce IL-4 against LACK antigen, resulting in resistance to leishmaniasis infection. The use of this protein as a DNA vaccine in mice was immunogenic and protective (32).
Another selective antigen is a cysteine protease involved in developing parasitic diseases and survival, host cell infection, and escaping from the host immune system; therefore, it has been suggested as a target therapy molecule. Cysteine proteases are considered pathogens of Leishmania parasites. Protein derived from cysteine protease B is involved in modulating immune system activities such as inducing IL-4 production, inhibiting the production of IL-12 by macrophages, and analyzing MHC-II molecules (33, 34).
Multiepitope peptide vaccines (Novel vaccines) designed by bioinformatics tools not only cope with a variety of pathogens effectively but also reduce the negative effects of irrelevant immune sequences (35). Despite all the benefits of using a multiepitope peptide vaccine, the prime issue with these vaccines is a low immune promotion (36). To solve the problem, adjuvants are recommended (37).
Toll-like receptors (TLRs) are protected receptors that are expressed in immune and non-immune cells and are known as a class of pattern recognition receptors (PRRs); those specific molecules recognize pathogens as agonists. Two TLR4 agonists, RpfE and RpfB, have been used as adjuvants to enhance vaccine safety. Various studies have used them as effective adjuvants in vaccines (38, 39).
Novel vaccines (Multiepitope peptide vaccines) include protected B and T cell epitopes, which can be a useful approach for the advancement of vaccines for infectious diseases. Studies have been performed to design multiepitope peptide vaccines for disease agents that have shown their ability to provide immunity against these agents (40-42).
We attempted to identify and anticipate the best and most successful functional B and T cell epitopes for immunization in this work using bioinformatics technologies. The resuscitation-promoting factor RpfE and RpfB of Mycobacterium TB are two TLR4 agonists that were used as adjuvants in this study. In addition, we focused on producing a recombinant multiepitope vaccine that contained effective antigenic epitopes.
The data illustrated the structure of the recombinant multiepitope vaccine was not allergic and could evoke humoral and cellular immune responses. The vaccine structure based on the bioinformatics system, which is reported here, provides significant immunogenic potential that may be further evaluated in the next phase of in vivo and in vitro study.


 

Materials and Methods

2.1. Protein Sequence Retrieval

Protein sequences of candidate antigens related to LACK (Accession no. AAB88300.1), CPB (Accession no. AUL80104.1), and KMP-11 (Accession no. AAR84616.1) were attained from the National Centre for Biotechnology Information        (https://www.ncbi.nlm.nih.gov/) (43).
Mycobacterium TB RpfE and RpfB were obtained from UniProt at www.uniprot.org and used as TLR4 agonists. RpfE (Entry: 053177) and RpfB (Entry: 053177) (Entry: P9WG29, G5 domain).

2.2. Evaluation of Antigen-determining Characteristics

ProtParam server was used for physicochemical characterization of 3 selected antigens like molecular weight, amino acid composition, half-life, aliphatic index, theoretical pI, GRAVY, and so on (https://web.expasy.org/protparam/) (44).
Furthermore, the antigenicity of every protein was shown using the ANTIGENpro server. This server (http://scratch.proteomics.ics.uci.edu/) is a pathogen -independent predictor with an accuracy of 82 percent. (45).
The SOPMA (Self-Optimized Prediction Method with Alignment) secondary structure analysis tool was used to predict four states: helix, beta-sheet, coil, and turn (https://npsa-prabi.ibcp.fr/NPSA/npsa_sopma.html) (46).

2.3. Anticipation of MHC-II Epitopes

MHC-II binding epitopes were selected through RANKPEP server. Human MHC-II alleles, which are HLA-DRB 1101, HLA-DRB 0401, and HLA-DRB 1 101 were selected to predict the MHC-II binding epitope.
RANKPEP server predicts MHCII epitopes based on position-specific scoring matrices (PSSMs). The binding threshold for the highest epitope scores is 4-6% MHC-II (http://imed.med.ucm.es/Tools/rankpep.html) (47).

2.4. Prediction of MHC-I Epitopes

The MHC-I epitope analysis was assessed using the IEDB server. HLA-A * 01: 01 and HLA-B * 07: 02 were among the alleles used to predict MHC-I binding. Artificial Neural Networks (ANN), Stabilized Matrix Approaches (SMM), and Score Matrices derived from Combinatorial Peptide Libraries may all be employed as estimation methods. The consensus technique (https://www.iedb.org/) is employed, a mix of multiple approaches like ANN, SMM, and other combine the algorithms (48, 49).

2.5. Prediction of B-Cell Epitopes

The BCpred server was applied for linear anticipation of B cell epitopes. The BCpred server uses a Subsequence String Kernel (SSK) and Support Vector Machine (SVM) with 74.54% and 75% accuracy and specificity, respectively    (https://webs.iiitd.edu.in/raghava/bcepred/) (50).

2.6. Prediction of CTL Epitopes

The CTLPred service was used to anticipate cytotoxic T lymphocyte (CTL) epitopes. This server employs a direct approach incorporating quantitative matrix (QM) and machine learning approaches. In addition, the server employs a mix of methodologies and consensus estimation. The consensus method technique was used to anticipate the default cut-off score (0.00) (http://www.webs.iiitd.edu.in/raghava/ctlpred/index.html). (51).

2.7. Anticipation of Interferon-Gamma Induction Epitopes

Interferon gamma-induced epitopes of MHC-II binding epitopes are predicted to design a useful multiepitope recombinant vaccine. The IFNepitope server uses a database made of IFN-gamma-induced and non-induced MHCII. IFNepitope server also uses various methods, including machine-learning, motif-based search, and a hybrid approach. The maximum accuracy attained from the hybrid model is 81.39% (https://webs.iiitd.edu.in/raghava/ifnepitope/index.php) (52).

2.8. Design of a Recombinant Vaccine

To create a recombinant multiepitope vaccine, the results obtained from the predicted epitopes were compared, and the epitopes with the highest and common scores were selected. Selected epitopes can evoke different sorts of immune responses. Selected epitopes were combined with adjuvants by a suitable linker.

2.9. Evaluation of the Characteristics of the Recombinant Multi-epitope Vaccine

The antigenic and antigenic responses of the recombinant vaccine construct were assessed using the Vaxijen v2.0 server. Vaxijen v2.0 server predicts antigenicity antigenic response based on target organism antigens such as bacterial, viral, and tumoral antigens.
This server employs a cutting-edge, alignment-independent method for predicting antigenic reaction probability. Its target organism accuracy ranges from 70% to 85% (http://www.ddgpharmfac.net/vaxijen/VaxiJen/VaxiJen.html) (53).
The AllerTop v.2.0 server was used to estimate the allergenicity of the recombinant vaccination. With an accuracy of 85.3 percent, this site predicts allergies and non-allergies using the K-Nearest Neighbors (KNN) algorithm           
(http://www.ddg-pharmfac.net/AllerTOP) (54).
The physicochemical characteristics of the recombinant multiepitope vaccination were assessed using a ProtParam server. In vitro and in vivo half-life, theoretical pI, molecular weight (MW), amino acid composition, instability index, in vitro and in vivo half-life, aliphatic index, and Grand average of hydropathicity index (GRAVY), and molecular weight (MW) were among the physicochemical parameters calculated (https://web.expasy.org/protparam/) (44).
2.10. Secondary Structure Evaluation
The server GOR IV, secondary structure (alpha-helix, beta-sheet, turn, or random coil) was analyzed (https://npsaprabi.ibcp.fr/cgi-bin/secpred_gor4.pl) (53).

2.11. Homogeneity Modeling

The third structure of the recombinant multiepitope vaccine was predicted by utilizing the I-TASSER server. The predicted three-dimensional models are known as confidence scores, known as C scores (higher values indicate higher confidence) (http://zhanglab.ccmb.med.umich.edu/I-TASSER/ download/) (54).

2.12. Refinement of 3D Model Structure

GalaxyRefine performed the refining process of the 3D model. According to C score, the best 3D model from I-TASSER has been processed in GalaxyRefine server. GalaxyRefine is likely to improve the early models (http://galaxy.seoklab.org/cgi-bin/submit.cgi?type=REFINE) (55).

2.13. Validation of the Refined Third Structure

The 3D manufacture of the ultimate vaccine has been verified using ProSAweb, Verify3D, Rampage, and ERRAT servers.
Using RAMPAGE's phi-psi torsion angles, RAMPAGE divides the protein into three groups: preferred outlier areas, permitted regions, and outlier regions. (http://mordred.bioc.cam.ac.uk/~rapper/rampage.php) (56).
ProSAweb calculates the total quality score for a particular input structure. The server simply needs Cα atoms to evaluate low-resolution structures. Highlighting errors in experimental and theoretical models of protein structures is a great issue in structural biology.
ProSA-web is a web-based interface to the ProSA software, which is often used to validate protein structures. For a given input structure, ProSA produces an overall quality score. If this rating falls outside of a typical range for natural proteins, the design is likely to have mistaken.      (https://prosa.services.came.sbg.ac.at/prosa.php) (57).
Non-bonded atomic interactions allow the ERRAT server to discriminate between proper and erroneous protein structure regions.               
(http://servi ces.mbi.ucla.edu) (58).
A three-dimensional protein model was tested for compliance with the vaccine's sequence using Verify3D to validate its three-dimensional structure. (http://services.mbi.ucla.edu/) (59).

2.14. In silico Cloning

The Sequence Manipulation Suite server (https://www.bioinformatics.org/sms/) was used to conduct reverse translation and codon optimization of vaccine protein sequences.
Another method used to investigate sequence properties was the GenScript Rare Codon Analysis Tool, which can be found at                https://www.genscript.com/tools/rare-codon-analysis  (https://www.genscript.com/tools/rare-codon-analysis) (CFD).
Eventually, restriction sites HindIII and BamHI were introduced to the N and C terminal sequences of the ultimate vaccine's DNA to clone it into the E. coli vector pET-14b.
 

 

Results

3.1. Retrieve and Collect Sequences

In the present study, LACK, CPB, and KMP-11 proteins were selected as antigenic determinants. Sequences for three antigenic peptides were retrieved in the FASTA format at the NCBI site. The amino acid sequences for RpfE and RpfB were obtained in FASTA format from the UniProt dataset. RpfB is the most important part of linker to TLR4 (60).

3.2. Analysis of Antigen-determining Structures

The physicochemical properties, secondary structure analysis, and antigenicity prediction of all three proteins were determined and summarized in Table 1.


Table 1. Analysis of selective antigens

LACK KMP-11 CPB Features evaluation of antigen determinants
312 92 349 Number of amino acids
34.42 KD 11.23 KD 37.95 KD Molecular weight
6.05 5.96 7.09 Theoretical pI
36 20 31 Total number of negatively charged residues (Asp + Glu)
30 17 31 Total number of positively charged residues (Arg + Lys)
>10 hours >10 hours >10 hours Estimated half-life (Escherichia coli, invivo)
31.38 50.02 35.65 Instability index
80.61 33.04 77.48 Aliphatic index
-0.275 -1.447 -0.095 Grand average of hydropathicity (GRAVY)
13 is   4.17% 77 is  83.70% 117 is  33.52% Alpha helix
136 is  43.59% 0 is   0.00% 63 is  18.05% Extended strand
47 is  15.06% 2 is   2.17% 19 is   5.44% Beta turn
116 is  37.18% 13 is  14.13% 150 is  42.98% Random coil
0.751214 0.456471 0.775114 Predicted Probability of Antigenicity

3.3. MHC Class II and I Epitopes

RANKPEP server used LACK, CPB, and KMP-11 proteins to anticipate MHC class II binding epitope (Tables 2). The MHC class I binding epitope anticipation was used by the server IEDB (Table 3).


Table 2. Details of MHC-II epitopes

Protein Name Start Position Allele Sequence Score
CPB 241 DRB 0101 YVSMESSER 20.861
303 DRB 0401 YWVIKNSWG 19.943
116 DRB 0101 YRKARADLS 18.08
104 DRB 0101 YFAAAKQHA 17.994
105 DRB 0401 FAAAKQHAG 17.737
251 DRB 1101 MAAWLAKNG 14.691
163 DRB 1101 WAVAGHKLV 10.425
KMP-11 70 DRB 1101 SEHFKQKFA 17.535
77 DRB 0101 FAELLEQQK 12.767
48 DRB 0401 YEKFERMIK 11.919
5 DRB 0101 YEEFSAKLD 9.643
LACK 248 DRB 0101 FWMCVATER 23.317
149 DRB 0401 HEDWVSSIC 20.452
41 DRB 1101 WKANPDRHS 18.624
26 DRB 0101 YIKVVLTSR 18.44
85 DRB 1101 WDRSIRMWD 16.17
10 DRB 0401 HRGWVTSLA 15.115


Table 3. Details of MHC-I epitopes

Protein Name Start Position Allele Sequence Score
CPB 339 HLA-A*01:01 HVSQSPTPY 0.41
265 HLA-A*01:01 VDASSFMSY 0.35
170 HLA-B*07:02 LVRLSEQQL 0.28
90 HLA-A*01:01 SEAEFAARY 0.23
73 HLA-B*07:02 QARNPHARF 0.23
118 HLA-B*07:02 KARADLSAV 0.19
KMP--11 40 HLA-A*01:01 LSPEMKEHY 0.35
69 HLA-A*01:01 HSEHFKQKF 0.24
34 HLA-B*07:02 KPDESTLSP 0.12
81 HLA-A*01:01 LEQQKAAQY 0.11
LACK 287 HLA-A*01:01 WSADGNTLY 0.96
253 HLA-A*01:01 ATERSLSVY 0.95
159 HLA-B*07:02 SPSLEHPIV 0.55
188 HLA-A*01:01 RTLKGHSNY 0.35
19 HLA-B*07:02 CPQQAGSYI 0.29
164 HLA-B*07:02 HPIVVSGSW 0.29
42 HLA-B*07:02 KANPDRHSV 0.28

 

3.4. Prediction of Interferon-gamma Induced Epitopes and B Cell Epitopes

The IFN epitope server was used to find the vaccine's IFN-gamma-inducing epitopes.
IFN gamma-inducing epitopes were categorized into 4 different epitopes. (Table 4).
BCpred server selected B-cell epitopes of three selected proteins. The results are shown in Table 5.


Table 4. IFN epitope server identified IFN-gamma-inducing epitopes in the ultimate vaccination design.

Protein Start–end position IFN gamma score Sequence
CPB 104-124 1.6909292 YFAAAKQHAGQHYRKARADLS
241-260 2 YVSMESSERVMAAWLAKNGP
KMP-11 48-78 4.3950929 YEKFERMIKEHTEKFNKKMHEHSEHFKQKF
LACK 26-49 3.3236104 YIKVVLTSRDGTAISWKANPDRHS


Table 5. Details of B-cell epitopes

Protein Name Start Position Sequence Score
CPB 209 FTEKSYPYVSGNGDVPECSN 0.988
300 EVPYWVIKNSWGKDWGEKGY 0.985
329 CLLTGYPVSVHVSQSPTPYL 0.978
126 VPDAVDWREKGAVTPVKNQG 0.977
KMP-11 31 FADKPDESTLSPEMKEHYEK 0.977
LACK 169 SGSWDNTIKVWNVNGGKCER 0.973
38 AISWKANPDRHSVDSDYGLP 0.931
191 KGHSNYVSTVTVSPDGSLCA 0.898

3.5. Prediction of CTL Epitopes

The CTLpred server detected high-ranking CTL epitopes.
When the MHC-I binding data were examined, researchers analyzed the parts that overlapped with the CTLPred results. Furthermore, CTL epitopes derived from frequent sites were used. (Table 6).

 
Table 6. Predicted CPB, KMP-11, and LACK protein CTL epitope.

Protein Name Start Position Sequence Score
CPB 63 ERNLELMRE 1.000
83 ITKFFDLSE 1.000
115 HYRKARADL 1.000
KMP-11 60 EKFNKKMHE 0.990
LACK 25 SYIKVVLTS 1.000
104 LKHTKDVLA 0.990

3.6. Selection of Final Epitopes and Development of a Vaccine with Many Epitopes

According to high-grade and common MHC-I, MHC-II, CTL, B cell, INF-gamma epitopes, 8 sections of 3 antigenic proteins were selected as the final region (Table 7). The GSGSGS linker fused selected epitopes of each protein. RpfE and RpfB were also combined with both ends of the vaccine as adjuvants. The final structure of the vaccine consists of 518 amino acid residues, shown in Figure 1.


Table 7. The eight final epitope segments were selected based on the findings of the various servers from three antigens

Protein Name Start–end position Sequence
CPB 63-92 ERNLELMREHQARNPHARFGITKFFDLSEA
104-124 YFAAAKQHAGQHYRKARADLS
219-260 GNGDVPECSNSSELAPGARIDGYVSMESSERVMAAWLAKNGP
300-345 EVPYWVIKNSWGKDWGEKGYVRVTMGVNACLLTGYPVSVHVSQSPT
KMP-11 48-78 YEKFERMIKEHTEKFNKKMHEHSEHFKQKFA
LACK 26-49 YIKVVLTSRDGTAISWKANPDRHS
149-187 HEDWVSSICFSPSLEHPIVVSGSWDNTIKVWNVNGGKCE
248-262 FWMCVATERSLSVYD


 Figure 1. A representation of vaccine sequence arrangement.
Figure 1. A representation of vaccine sequence arrangement.

 

3.7. Allergenicity and Antigenicity Evaluation

The result retrieved by the AllerTOP 2.0 server showed that the designed structure is non-allergenic.
As a result, the multiepitope vaccine has been created such that this will not generate allergy-specific IgE and inflammation.
The vaccine has a total antigenocytic probability of 0.8659 percent by Vaxijen at a minimum of 0.4 percent, indicating that it may induce effective T and B cell immune function.

3.8. Measurement of Physicochemical Parameters

According to our calculations, the protein's theoretical pI is 5.87, and its molecular weight is 55.05 kDa. The pI value is a measure of the protein's ph. Remainders with a total of 61 negative and 52 positive charges were found in this study. The anticipated half-lives of mammalian erythrocytes (in vitro), yeast (in vivo), and bacteria (in vitro) were 100, 20, and 10 hours, respectively (E. coli, in vivo). It was determined that the instability index (II) was 37.70. Stable protein is what we call this one. 61.58 was the aliphatic index value. The protein has a high aliphatic index, implying it can withstand a broad temperature. Aside from that, the vaccine construct's GRAVY value was -0.506. Negative GRAVY implies that the protein is hydrophilic and more capable of interacting with water molecules in the environment.

3.9. Secondary Structure Evaluation

Secondary structure prediction by GOR IV showed that the multi-epitope vaccine includes 24.13% α-helix (H), 20.66% extended strand (E), 0.00% beta-turn (T) and 55.21% random coil (C) (figure 2). The proportion of secondary structures in the multiepitope structure suggests that the designed vaccine is likely to be able to form antigenic epitopes.

 Figure 2. The predicted secondary structure of the multiepitope vaccines using GOR IV software. H: Alpha helix (blue), E: Extended strand (red), T: Beta-turn (green) and C: Random coil (yellow)

 Figure 2. The predicted secondary structure of the multiepitope vaccines using GOR IV software. H: Alpha helix (blue), E: Extended strand (red), T: Beta-turn (green) and C: Random coil (yellow)  Figure 2. The predicted secondary structure of the multiepitope vaccines using GOR IV software. H: Alpha helix (blue), E: Extended strand (red), T: Beta-turn (green) and C: Random coil (yellow)

Figure 2. The predicted secondary structure of the multiepitope vaccines using GOR IV software. H: Alpha helix (blue), E: Extended strand (red), T: Beta-turn (green) and C: Random coil (yellow)
 

3.10. Homogeneity Modeling

The I-TASSER server was used to create the final vaccine's 3D model, and the best five approaches rely upon C score were presented. The C score ranges from [-5 to 2], and the greater the C score, the more serious the problem, the more confident the model is. Model 3 had the best C-score (-1.76) and was chosen for more study. (Figure 3a).

3.11. Refinement of the Structure of the Third Model

Selected models were refined by applying GalaxyRefine. Five improved 3D models were released to the GalaxyRefine service. Furthermore, the best model was obtained based on the Z score and the overall quality coefficient of a high-quality three-dimensional model (Figure 3b).

 
Figure 3. The refined 3D model of the multiepitope peptide vaccine. A) The 3D structure of the designed vaccine was determined by homology modeling using I-TASSER server, and B) was refined by GalaxyRefine and 3Drefine servers. 
Figure 3. The refined 3D model of the multiepitope peptide vaccine. A) The 3D structure of the designed vaccine was determined by homology modeling using I-TASSER server, and B) was refined by GalaxyRefine and 3Drefine servers.
 

3.12. Validation of the Refined Third Structure

Many virtual validating techniques were utilized to check the validity of the revised model. These approaches included: Ramachandran design; ERRAT; ProSA; and verify-3D.
For example, 337 (79.858 %), 56 (13.270 percent), and 29 (6.872 %) of the original model residues were found in the preferred, permitted, and outlier areas, respectively (Figure 4a).
393 (93.128 %), 18 (4.265 %), and 11 (2.607 percent) were the new ratios after the refining run (Figure 4b).
It seems from the findings that most of the amino acids have been moved inside the permitted range throughout the refining run, according to the data.

Figure 4. The validation of 3D protein model, using Ramachandran plot. a) The initial model b) The refined model
Figure 4. The validation of 3D protein model, using Ramachandran plot. a) The initial model b) The refined model

 

ProSA-web server assessments were also included in our research to further confirm the quality of the 3D model before and after refining. The original model's ProSA Z-score was -1.73, while the improved model's Z-score was -3.36 (Figure 5a and 5b, respectively). Structures of comparable size to natural proteins have been included in this model, as seen in Figure 5. It was also shown that most residues had negative energy values when plotted on a graph based on energy. (Figure 6).


Figure 5. The z-Score graphs for the construct's 3D structure. 
Figure 5. The z-Score graphs for the construct's 3D structure.

 

The first model's z-score is -1.73, which is beyond the range of natural protein structure, and b
After refining, a model's z-score is – 3.63, which is within the range of natural protein structure.
The z-Score graphic shows the z-scores of all experimentally determined polypeptide chains in PDB (dark blue) and X-ray crystallography (light blue) (light blue).
Results with a z-score of 10 are shown in the graph.
The protein's z score is shown by a huge black dot.

 
Figure 6. ProSA server energy charts of the original model (a) and revised model (b). For the improved model, the majority of the residues have negative numbers, as illustrated.  
Figure 6. ProSA server energy charts of the original model (a) and revised model (b). For the improved model, the majority of the residues have negative numbers, as illustrated.

 

In addition, the quality of the modeled structure was verified using ERRAT. The results showed that the overall quality coefficient of the initial three-dimensional model was 58.5106 (Figure 7a). After refining processes, the ERRAT coefficient of the refined 3D model reached 73.4818 (Figure 7b).

 
Figure 7. The overall quality factor plot (ERRAT) of a the initial model is 58.5106, and b the final model after adjustment is 73.4818; areas of the 3D model that could be refused at the 99 percent confidence level are shown with grey lines, and zones that can be rejected at the 95 percent confidence level have been shown with black lines in the ERRAT graph. The total qualityof f value for excellent high-resolution structures is about 95% or above.  
Figure 7. The overall quality factor plot (ERRAT) of a the initial model is 58.5106, and b the final model after adjustment is 73.4818; areas of the 3D model that could be refused at the 99 percent confidence level are shown with grey lines, and zones that can be rejected at the 95 percent confidence level have been shown with black lines in the ERRAT graph. The total qualityof f value for excellent high-resolution structures is about 95% or above.

 

Furthermore, the Verify 3D score revealed that 53.86 percent of the residues in the original model had a mean score of 3D-1D score 0.2. (Figure 8a). The Verify 3D score was 58.69 percent after the refining procedure, showing that further residues were put in appropriate side chain settings. (Figure 8b).

 
Figure 8. The Verify-3D program evaluates the quality of 3D buildings. In the original model (a) and refined model (b), 53.86 percent and 58.69 percent of the residues, respectively, have a score of > 0.2. 
Figure 8. The Verify-3D program evaluates the quality of 3D buildings. In the original model (a) and refined model (b), 53.86 percent and 58.69 percent of the residues, respectively, have a score of > 0.2.

 

3.13. In silico Cloning

The vaccine construct was developed by the Sequence Manipulation Suite server was backward translated and codon-optimized. The GenScript program was used to assess crucial gene sequence properties such as codon compatibility index (CAI), GC content, and codon frequency distribution in order to enable high-level protein production in E. coli hosts (CFD). CAI was the optimized nucleotide sequence 1, CAI> 0.80 is considered suitable for expression. The mean GC content of the designed vaccine sequence was 59.37%; the percentage of GC content in the range of 30-70% is desirable. The frequency distribution of the codon was 100 (CFD), while codons with values <30 appeared to be inhibited. Overall, these findings indicated that the optimized DNA sequence is clonable and expression-ready.

 
 

Discussion

Leishmaniasis is a widespread parasitic disease that is prevalent in most regions of the globe. It may take many different forms, given the variety of pathogenic organisms. (61).
Leishmania major is one of the causes of occurring cutaneous leishmaniasis, which is more common in rural areas with poor health conditions and low economic status. Leishmania major can cause skin lesions. These lesions can spread and become painful. Tragically, even after healing, the lesion stays distorted, and the skin is injured in most instances. (62).
Various factors, including the unavailability of the vaccine, proper control of the vector, the cost of available drugs, the duration of treatment, and resistance to the parasite, all play a role in the inability to eradicate the disease completely. Therefore, the best strategy is to produce an effective vaccine that can stimulate and activate the body's immune system against the parasite (63, 64).
Basic studies are needed to identify a safe and effective vaccine that can stimulate the immune response (65). Antigens such as CP and KMP-11 have been investigated as targets in vaccine design for combat Leishmania (66, 67). In addition, recombinant vaccines such as Leish111f and Leishmune have been developed as leishmaniasis vaccination alternatives (68). Yet, no ideal vaccination against the illness has been authorized for human treatment (13, 69).
Bioinformatics approaches are now effective for designing novel vaccines (70). Several important studies have demonstrated the benefits of vaccines that have been designed using bioinformatics methods and are currently used as an effective vaccine (e.g., studies on malaria, influenza, cancer, dengue, and multiple sclerosis) (19, 71-73).
Several bioinformatics and immunoinformatics techniques have been effectively applied in numerous biological disciplines. These tools reduce the time and cost required to identify the dominant immune epitopes of B and T cells while increasing the level of screening accuracy (74).
Research shows that Leishmania major infection in a resistant mouse model; it activates Th1 type CD4 cells. Th1 cells produce interferon-gamma, which promotes improvement and immunity against infection. The Th2 response is induced in BALB/c sensitive mice, and IL-4 is generated; however, IFN- production is decreased, and infected animals are susceptible to the illness. However, in the mouse model, the production of the Th1 response type is linked to therapy and protection, while the generation of the Th2 response type is linked to disease progression and mortality, but human leishmaniasis susceptibility and resistance are yet unknown (20, 75-78).
Overall, the recovery form of CL is associated with cells that generate IFN-, while the non-recovery form of CL is associated with a combination of Th1 and Th2 cytokines, with IL-4 and IL-10 being abundant (79-81). As a result, vaccinations targeting T and B cell epitopes seem to be more effective. As a result, T and B cell epitopes were investigated in this research.
Employing bioinformatics techniques, the antigens LACK, CPB, and KMP-11 were utilized to construct a multiepitope vaccination for L. major. In order to enhance the likelihood of discovering the greatest immunodominant epitopes, numerous servers were used to choose LACK, CPB, and KMP-11 antigens. MHC-I, MHC-II, CTL, B cell, and INF-gamma binding epitopes were compared, and epitopes with high scores and overlaps were chosen. In addition to predicting T and B cell epitopes, the presence of IFN-γ-induced epitopes in the structure of the ultimate vaccine was evaluated. Four epitopes were identified as IFN-γ-induced epitopes. Several cytokines, notably IFN-, have been demonstrated to be significant in parasite elimination. (82). Hence, epitopes capable of producing IFN-γ are important for CL vaccine design.
Adjuvants are added to the structure of multiepitope peptide vaccines to overcome the poor immunogenicity of the vaccine. Adjuvants play a crucial role in boosting the immune system (41, 83). Pathogen-associated molecular patterns (PAMPs) attach to Toll-like receptors (TLRs), which trigger innate immune responses. Two TLR 4 agonists, RpfB and RpfE, were utilized as adjuvants in this investigation. RpfB enhances Th1-type T cell immunological responses by directly binding to TLR4 and activating TLR4-dependent dendritic cells. RpfE interacts with DC to differentiate crude CD4 cells into Th1 and Th17 immune responses (62). As a result, using adjuvants to develop effective multiepitope vaccinations to prevent CL is a good idea.
Linkers are essential components of recombinant multiepitope vaccines and take part in functional structural vaccine development (79, 84). In this study, two types of linkers, namely EAAAK and GSGSGS, were used to join different parts of the multiepitope vaccine. The GSGSGS flexible linker was used to match functional domains that require domain interactions. EAAAK rigid linker creates space between domains (80, 81). In addition, these linkers provide stability and bioactivity to the peptide structure (83, 85). Accordingly, EAAAK was used to link RpfB and RpfE to the designed vaccine structure.
Bioinformatics tools evaluated the vaccine's physicochemical, immunological, and structural characteristics. According to the results of structural, immunological, and physicochemical properties, the vaccine designed in this study can be proposed as a suitable option for a vaccine.
Numerous factors, including CAI, CFD, and GC content, must be tuned to achieve high-level protein expression in E. coli. The findings of the optimized gene demonstrated that the intended vaccine could be efficiently produced in the E. coli host following analyzing all of the aforementioned parameters employing GenScript.


 

Conclusion

Using an immunoinformatics approach, we aimed to construct a multiepitope-based vaccination against CL in this work. Our created vaccine may be a decent choice for vaccination against Leishmania major based on computational findings, immunological and structural analyses, and physicochemical assessments. The recombinant structure can activate both humoral and cellular immune responses. The suggested vaccine, which uses a variety of epitopes and adjuvants and has acceptable physicochemical properties, is likely to elicit strong immune responses against CL. However, in vitro and in vivo testing are required to assess the effectiveness of multiple epitope vaccines.

 

Acknowledgment

The authors appreciate the Deputy of Research and Technology, Lorestan University of Medical Sciences, Khorramabad, Iran. This article is derived from the Master's thesis of the first author, Department of Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
 
 

Ethical Approval

The present study was approved by The Ethics Committee of Lorestan University of Medical Sciences (IR.LUMS.REC.1398.190).
 

 

Funding

This research was financially supported by Lorestan University of Medical Sciences, Khorramabad, Iran. Hereby the authors appreciate all the people who helped in this research.

 

Conflicts of Interest

The authors declare that there is no conflict of interest.


 

Type of Study: Original Research Article | Subject: Microbial Bioinformatics
Received: 2021/12/25 | Accepted: 2022/06/4 | ePublished: 2022/08/8

References
1. Kevric I, Cappel MA, Keeling JH. New world and old world Leishmania infections: a practical review. Dermatol Clin. 2015;33(3):579-93. [DOI:10.1016/j.det.2015.03.018] [PMID]
2. Kayani B, Sadiq S, Rashid HB, Ahmed N, Mahmood A, Khaliq MS, et al. Cutaneous Leishmaniasis in Pakistan: a neglected disease needing one health strategy. BMC Infect Dis. 2021;21(1):1-10. [DOI:10.1186/s12879-021-06327-w] [PMID] [PMCID]
3. Seyed N, Taheri T, Vauchy C, Dosset M, Godet Y, Eslamifar A, et al. Immunogenicity evaluation of a rationally designed polytope construct encoding HLA-A* 0201 restricted epitopes derived from Leishmania major related proteins in HLA-A2/DR1 transgenic mice: steps toward polytope vaccine. PLoS One. 2014;9(10): e108848. [DOI:10.1371/journal.pone.0108848] [PMID] [PMCID]
4. Miramin-Mohammadi A, Javadi A, Eskandari SE, Mortazavi H, Rostami MN, Khamesipour A. Immune response in cutaneous leishmaniasis patients with healing vs. non-healing lesions. Iran J Microbiol. 2020;12(3):249-55. [DOI:10.18502/ijm.v12i3.3243] [PMID] [PMCID]
5. Mousavi P, Rahimi Esboei B, Pourhajibagher M, Fakhar M, Shahmoradi Z, Hejazi SH, et al. Anti-leishmanial effects of resveratrol and resveratrol nanoemulsion on Leishmania major. BMC Microbiol. 2022;22(1):1-14. [DOI:10.1186/s12866-022-02455-8] [PMID] [PMCID]
6. Ezra N, Ochoa MT, Craft N. Human immunodeficiency virus and leishmaniasis. J Glob Infect Dis. 2010;2(3):248. [DOI:10.4103/0974-777X.68528] [PMID] [PMCID]
7. Lockard RD, Wilson ME, Rodríguez NE. Sex-Related Differences in Immune Response and Symptomatic Manifestations to Infection with Leishmania Species. J Immunol Res. 2019; 2019:4103819. [DOI:10.1155/2019/4103819] [PMID] [PMCID]
8. Alvar J, Vélez ID, Bern C, Herrero M, Desjeux P, Cano J, et al. Leishmaniasis worldwide and global estimates of its incidence. PloS one. 2012; 7(5):e35671. [DOI:10.1371/journal.pone.0035671] [PMID] [PMCID]
9. Global Burden of Disease Study C. Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;386(9995):743-800. [DOI:10.1016/S0140-6736(15)60692-4]
10. Madusanka RK, Silva H, Karunaweera ND. Treatment of Cutaneous Leishmaniasis and Insights into Species-Specific Responses: A Narrative Review. Infect Dis Ther. 2022:1-17. [DOI:10.1007/s40121-022-00602-2] [PMID] [PMCID]
11. Aoun K, Kalboussi Y, Sghaier IB, Souissi O, Hammami H, Bellali H, et al. Assessment of Incubation Period of Cutaneous Leishmaniasis due to Leishmania major in Tunisia. Am J Trop Med Hyg. 2020;103(5):1934-7. [DOI:10.4269/ajtmh.20-0439] [PMID] [PMCID]
12. Alghounaim M, Chivinski J, Barkati S. Cutaneous leishmaniasis in a 12-year-old Syrian immigrant. CMAJ. 2022;194(3):E93-E4. [DOI:10.1503/cmaj.210847] [PMID] [PMCID]
13. Rabienia M, Roudbari Z, Ghanbariasad A, Abdollahi A, Mohammadi E, Mortazavidehkordi N, et al. Exploring membrane proteins of Leishmania major to design a new multiepitope vaccine using immunoinformatics approach. Eur J Pharm Sci. 2020;152:105423. [DOI:10.1016/j.ejps.2020.105423] [PMID]
14. Erber AC, Sandler PJ, de Avelar DM, Swoboda I, Cota G, Walochnik J. Diagnosis of visceral and cutaneous leishmaniasis using loop-mediated isothermal amplification (LAMP) protocols: a systematic review and meta-analysis. Parasites Vectors. 2022;15(1):1-16. [DOI:10.1186/s13071-021-05133-2] [PMID] [PMCID]
15. Shams M, Nourmohammadi H, Basati G, Adhami G, Majidiani H, Azizi E. Leishmanolysin gp63: Bioinformatics evidences of immunogenic epitopes in Leishmania major for enhanced vaccine design against zoonotic cutaneous leishmaniasis. Inform Med Unlocked. 2021;24: 100626. [DOI:10.1016/j.imu.2021.100626]
16. Kedzierski L. Leishmaniasis vaccine: where are we today? J Glob Infect Dis. 2010;2(2):177. [DOI:10.4103/0974-777X.62881] [PMID] [PMCID]
17. Vélez ID, Gilchrist K, Martínez S, Ramírez-Pineda JR, Ashman JA, Alves FP, et al. Safety and immunogenicity of a defined vaccine for the prevention of cutaneous leishmaniasis. Vaccine. 2009;28(2):329-37. [DOI:10.1016/j.vaccine.2009.10.045] [PMID]
18. Okwor I, Mou Z, Dong L, UZONNA JE. Protective immunity and vaccination against cutaneous leishmaniasis. Front Immunol. 2012;3:128. [DOI:10.3389/fimmu.2012.00128] [PMID] [PMCID]
19. Adu-Bobie J, Capecchi B, Serruto D, Rappuoli R, Pizza M. Two years into reverse vaccinology. Vaccine. 2003;21(7-8):605-10. [DOI:10.1016/S0264-410X(02)00566-2]
20. Sacks D, Noben-Trauth N. The immunology of susceptibility and resistance to Leishmania major in mice. Nat Rev Immunol. 2002;2(11):845-58. [DOI:10.1038/nri933] [PMID]
21. Lakhal-Naouar I, Koles N, Rao M, Morrison EB, Childs JM, Alving CR, et al. Transcutaneous immunization using SLA or rLACK skews the immune response towards a Th1 profile but fails to protect BALB/c mice against a Leishmania major challenge. Vaccine. 2019;37(3):516-23. [DOI:10.1016/j.vaccine.2018.11.052] [PMID]
22. Dariushnejad H, Ghorbanzadeh V, Akbari S, Hashemzadeh P. Design of a Novel Recombinant Multi-Epitope Vaccine against Triple-Negative Breast Cancer. Iran Biomed J. 2022;26(2):160-74.
23. Dariushnejad H, Ghorbanzadeh V, Hashemzadeh P. Prediction of B-and T-cell epitopes using in-silico approaches: a solution to the development of recombinant vaccines against covid-19. Minerva Biotechnol Biomol Res. 2021;33(1):36-42. [DOI:10.23736/S2724-542X.20.02652-X]
24. Chen H-Z, Tang L-L, Yu X-L, Zhou J, Chang Y-F, Wu X. Bioinformatics analysis of epitope-based vaccine design against the novel SARS-CoV-2. Infect Dis Poverty. 2020;9(1):1-10. [DOI:10.1186/s40249-020-00713-3] [PMID] [PMCID]
25. Foroutan M, Ghaffarifar F, Sharifi Z, Dalimi A, Pirestani M. Bioinformatics analysis of ROP8 protein to improve vaccine design against Toxoplasma gondii. Infect Genet Evol. 2018;62: 193-204. [DOI:10.1016/j.meegid.2018.04.033] [PMID]
26. Hashemzadeh P, Rouzbahani Ak, Bandehpour M, Kheirandish F, Dariushnejad H, Mohamadi M. Designing a recombinant multiepitope vaccine against Leishmania donovani based immuno-informatics approaches. Minerva Biotecnol. 2020;32(2):52-7. [DOI:10.23736/S1120-4826.20.02610-5]
27. Marciani DJ. Vaccine adjuvants: role and mechanisms of action in vaccine immunogenicity. Drug Discov Today. 2003;8(20): 934-43. [DOI:10.1016/S1359-6446(03)02864-2]
28. Liljeroos L, Malito E, Ferlenghi I, Bottomley MJ. Structural and computational biology in the design of immunogenic vaccine antigens. J Immunol Res. 2015;2015. [DOI:10.1155/2015/156241] [PMID] [PMCID]
29. Todolí F, Solano-Gallego L, De Juan R, Morell P, del Carmen Núñez M, Lasa R, et al. Humoral and in vivo cellular immunity against the raw insect-derived recombinant Leishmania infantum antigens KMPII, TRYP, LACK, and papLe22 in dogs from an endemic area. Am J Trop Med Hyg. 2010; 83(6):1287-94. [DOI:10.4269/ajtmh.2010.09-0784] [PMID] [PMCID]
30. Russo D, Turco S, Burns J, Reed S. Stimulation of human T lymphocytes by Leishmania lipopho-sphoglycan-associated proteins. J Immunol. 1992;148(1):202-7.
31. Mortazavidehkordi N, Fallah A, Abdollahi A, Kia V, Khanahmad H, Najafabadi ZG, et al. A lentiviral vaccine expressing KMP11-HASPB fusion protein increases immune response to Leishmania major in BALB/C. Parasitol Res. 2018;117(7):2265-73. [DOI:10.1007/s00436-018-5915-6] [PMID]
32. Rodríguez-Cortés A, Ojeda A, López-Fuertes L, Timón M, Altet L, Solano-Gallego L, et al. Vaccination with plasmid DNA encoding KMPII, TRYP, LACK and GP63 does not protect dogs against Leishmania infantum experimental challenge. Vaccine. 2007;25(46):7962-71. [DOI:10.1016/j.vaccine.2007.08.023] [PMID]
33. Banuls A-L, Hide M, Prugnolle F. Leishmania and the leishmaniases: a parasite genetic update and advances in taxonomy, epidemiology and pathogenicity in humans. Adv Parasitol. 2007;64: 1-458. [DOI:10.1016/S0065-308X(06)64001-3]
34. Mottram JC, Coombs GH, Alexander J. Cysteine peptidases as virulence factors of Leishmania. Curr Opin Microbiol. 2004;7(4):375-81. [DOI:10.1016/j.mib.2004.06.010] [PMID]
35. He J, Huang F, Li J, Chen Q, Chen D, Chen J. Bioinformatics analysis of four proteins of Leishmania donovani to guide epitopes vaccine design and drug targets selection. Acta tropica. 2019;191:50-9. [DOI:10.1016/j.actatropica.2018.12.035] [PMID]
36. Joshi S, Rawat K, Yadav NK, Kumar V, Siddiqi MI, Dube A. Visceral leishmaniasis: advancements in vaccine development via classical and molecular approaches. Front Immunol. 2014;5:380. [DOI:10.3389/fimmu.2014.00380] [PMID] [PMCID]
37. Lee S, Nguyen MT. Recent advances of vaccine adjuvants for infectious diseases. Immune Netw. 2015;15(2):51-7. [DOI:10.4110/in.2015.15.2.51] [PMID] [PMCID]
38. Reed SG, Hsu F-C, Carter D, Orr MT. The science of vaccine adjuvants: advances in TLR4 ligand adjuvants. Curr Opin Immunol. 2016;41:85-90. [DOI:10.1016/j.coi.2016.06.007] [PMID]
39. Black M, Trent A, Tirrell M, Olive C. Advances in the design and delivery of peptide subunit vaccines with a focus on toll-like receptor agonists. Expert rev vaccines. 2010;9(2):157-73. [DOI:10.1586/erv.09.160] [PMID] [PMCID]
40. Hashemzadeh P, Ghorbanzadeh V, Valizadeh Otaghsara SM, Dariushnejad H. Novel predicted B-cell epitopes of PSMA for development of prostate cancer vaccine. Int J Pept Res Ther. 2020;26(3):1523-5. [DOI:10.1007/s10989-019-09954-9]
41. Hashemzadeh P, Ghorbanzadeh V, Lashgarian HE, Kheirandish F, Dariushnejad H. Harnessing Bioinformatic Approaches to Design Novel Multi-epitope Subunit Vaccine Against Leishmania infantum. Int J Pept Res Ther. 2019:1-12. [DOI:10.1007/s10989-019-09949-6]
42. Dariushnejad H, Ghorbanzadeh V, Akbari S, Hashemzadeh P. Designing a Multiepitope Peptide Vaccine against COVID-19 Variants Utilizing In-silico Tools. Iran J Med Microbiol. 2021;15(5):7. [DOI:10.30699/ijmm.15.5.592]
43. Pruitt KD, Tatusova T, Maglott DR. NCBI reference sequences (RefSeq): a curated non-redundant sequence database of genomes, transcripts and proteins. Nucleic Acids Res. 2007; 35(suppl_1):D61-D5. [DOI:10.1093/nar/gkl842] [PMID] [PMCID]
44. Gasteiger E, Hoogland C, Gattiker A, Wilkins MR, Appel RD, Bairoch A. Protein identification and analysis tools on the ExPASy server. The proteomics protocols handbook: Springer; 2005. p. 571-607. [DOI:10.1385/1-59259-890-0:571]
45. Magnan CN, Zeller M, Kayala MA, Vigil A, Randall A, Felgner PL, et al. High-throughput prediction of protein antigenicity using protein microarray data. Bioinformatics. 2010;26(23):2936-43. [DOI:10.1093/bioinformatics/btq551] [PMID] [PMCID]
46. Geourjon C, Deleage G. SOPMA: significant improvements in protein secondary structure prediction by consensus prediction from multiple alignments. Bioinformatics. 1995;11(6):681-4. [DOI:10.1093/bioinformatics/11.6.681] [PMID]
47. Reche PA, Glutting J-P, Zhang H, Reinherz EL. Enhancement to the RANKPEP resource for the prediction of peptide binding to MHC molecules using profiles. Immunogenetics. 2004;56(6):405-19. [DOI:10.1007/s00251-004-0709-7] [PMID]
48. Andreatta M, Nielsen M. Gapped sequence alignment using artificial neural networks: application to the MHC class I system. Bioinformatics. 2016;32(4):511-7. [DOI:10.1093/bioinformatics/btv639] [PMID] [PMCID]
49. Dhanda SK, Mahajan S, Paul S, Yan Z, Kim H, Jespersen MC, et al. IEDB-AR: immune epitope database-analysis resource in 2019. Nucleic Acids Res. 2019;47(W1):W502-W6. [DOI:10.1093/nar/gkz452] [PMID] [PMCID]
50. Chen J, Liu H, Yang J, Chou K-C. Prediction of linear B-cell epitopes using amino acid pair antigenicity scale. Amino acids. 2007;33(3):423-8. [DOI:10.1007/s00726-006-0485-9] [PMID]
51. 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]
52. Dhanda SK, Vir P, Raghava GP. Designing of interferon-gamma inducing MHC class-II binders. Biology direct. 2013;8(1):30. [DOI:10.1186/1745-6150-8-30] [PMID] [PMCID]
53. Xia F, Dou Y, Lei G, Tan Y. FPGA accelerator for protein secondary structure prediction based on the GOR algorithm. BMC Bioinform. 2011;12(1): 1-9. [DOI:10.1186/1471-2105-12-S1-S5] [PMID] [PMCID]
54. Yang J, Yan R, Roy A, Xu D, Poisson J, Zhang Y. The I-TASSER Suite: protein structure and function prediction. Nat Methods. 2015;12(1):7-8. [DOI:10.1038/nmeth.3213] [PMID] [PMCID]
55. Heo L, Park H, Seok C. GalaxyRefine: protein structure refinement driven by side-chain repacking. Nucleic Acids Res. 2013;41(W1): W384-W8. [DOI:10.1093/nar/gkt458] [PMID] [PMCID]
56. Lovell SC, Davis IW, Arendall III WB, De Bakker PI, Word JM, Prisant MG, et al. Structure validation by Cα geometry: ϕ, ψ and Cβ deviation. Proteins: Struct Funct Genet. 2003;50(3):437-50. [DOI:10.1002/prot.10286] [PMID]
57. Wiederstein M, Sippl MJ. ProSA-web: interactive web service for the recognition of errors in three-dimensional structures of proteins. Nucleic Acids Res. 2007;35(suppl_2):W407-W10. [DOI:10.1093/nar/gkm290] [PMID] [PMCID]
58. Colovos C, Yeates TO. Verification of protein structures: patterns of nonbonded atomic interactions. Protein Sci. 1993;2(9):1511-9. [DOI:10.1002/pro.5560020916] [PMID] [PMCID]
59. Eisenberg D, Lüthy R, Bowie JU. [20] VERIFY3D: assessment of protein models with three-dimensional profiles. Meth Enzymol. 277: Elsevier; 1997. p. 396-404. [DOI:10.1016/S0076-6879(97)77022-8]
60. Kim JS, Kim WS, Choi HG, Jang B, Lee K, Park JH, et al. Mycobacterium tuberculosis RpfB drives Th1‐type T cell immunity via a TLR4‐dependent activation of dendritic cells. J Leukoc Biol. 2013;94(4):733-49. [DOI:10.1189/jlb.0912435] [PMID]
61. Depledge DP, MacLean LM, Hodgkinson MR, Smith BA, Jackson AP, Ma S, et al. Leishmania-specific surface antigens show sub-genus sequence variation and immune recognition. PLoS Negl Trop Dis. 2010;4(9):e829. [DOI:10.1371/journal.pntd.0000829] [PMID] [PMCID]
62. Reithinger R, Dujardin J-C. Molecular diagnosis of leishmaniasis: current status and future applications. J Clin Microbiol. 2007;45(1):21-5. [DOI:10.1128/JCM.02029-06] [PMID] [PMCID]
63. Zahedifard F, Lee H, No JH, Salimi M, Seyed N, Asoodeh A, et al. Anti-leishmanial activity of Brevinin 2R and its Lauric acid conjugate type against L. major: In vitro mechanism of actions and in vivo treatment potentials. PLoS Negl Trop Dis. 2019;13(2):e0007217. [DOI:10.1371/journal.pntd.0007217] [PMID] [PMCID]
64. Khatoon N, Pandey RK, Prajapati VK. Exploring Leishmania secretory proteins to design B and T cell multiepitope subunit vaccine using immunoinformatics approach. Sci Rep. 2017; 7(1):1-12. [DOI:10.1038/s41598-017-08842-w] [PMID] [PMCID]
65. Moafi M, Rezvan H, Sherkat R, Taleban R. Leishmania vaccines entered in clinical trials: A review of literature. Int J Prev Med. 2019;10. [DOI:10.4103/ijpvm.IJPVM_116_18] [PMID] [PMCID]
66. Das A, Ali N. Combining cationic liposomal delivery with MPL-TDM for cysteine protease cocktail vaccination against Leishmania donovani: evidence for antigen synergy and protection. PLoS Negl Trop Dis. 2014;8(8):e3091. [DOI:10.1371/journal.pntd.0003091] [PMID] [PMCID]
67. Basu R, Bhaumik S, Basu JM, Naskar K, De T, Roy S. Kinetoplastid membrane protein-11 DNA vaccination induces complete protection against both pentavalent antimonial-sensitive and-resistant strains of Leishmania donovani that correlates with inducible nitric oxide synthase activity and IL-4 generation: evidence for mixed Th1-and Th2-like responses in visceral leishmaniasis. J Immunol. 2005;174(11):7160-71. [DOI:10.4049/jimmunol.174.11.7160] [PMID]
68. Jain K, Jain N. Vaccines for visceral leishmaniasis: A review. J Immunol Methods. 2015;422:1-12. [DOI:10.1016/j.jim.2015.03.017] [PMID]
69. Lari A, Lari N, Biabangard A. Immunoinformatics Approach to Design a Novel Subunit Vaccine Against Visceral Leishmaniasis. Int J Pept Res Ther. 2022;28(1):1-14. [DOI:10.1007/s10989-021-10344-3] [PMID] [PMCID]
70. Khatoon N, Ojha R, Mishra A, Prajapati VK. Examination of antigenic proteins of Trypanosoma cruzi to fabricate an epitope-based subunit vaccine by exploiting epitope mapping mechanism. Vaccine. 2018;36(42):6290-300. [DOI:10.1016/j.vaccine.2018.09.004] [PMID]
71. Kalita P, Padhi A, Zhang KY, Tripathi T. Design of a peptide-based subunit vaccine against novel coronavirus SARS-CoV-2. Microb Pathog. 2020:104236. [DOI:10.1016/j.micpath.2020.104236] [PMID] [PMCID]
72. Zheng J, Lin X, Wang X, Zheng L, Lan S, Jin S, et al. In silico analysis of epitope-based vaccine candidates against hepatitis B virus polymerase protein. Viruses. 2017;9(5):112. [DOI:10.3390/v9050112] [PMID] [PMCID]
73. Delany I, Rappuoli R, Seib KL. Vaccines, reverse vaccinology, and bacterial pathogenesis. Cold Spring Harb perspect med. 2013;3(5):a012476. [DOI:10.1101/cshperspect.a012476] [PMID] [PMCID]
74. Shahbazi M, Haghkhah M, Rahbar MR, Nezafat N, Ghasemi Y. In silico sub-unit hexavalent peptide vaccine against an Staphylococcus aureus biofilm-related infection. Int J Pept Res Ther. 2016;22(1):101-17. [DOI:10.1007/s10989-015-9489-1]
75. Scott P, Novais FO. Cutaneous leishmaniasis: immune responses in protection and pathogenesis. Nat Rev Immunol. 2016;16(9):581-92. [DOI:10.1038/nri.2016.72] [PMID]
76. Kemp K. Cytokine-producing T cell subsets in human leishmaniasis. Arch Immunol Ther Exp. 2000;48(3):173-6.
77. Modolell M, Choi B-S, Ryan RO, Hancock M, Titus RG, Abebe T, et al. Local suppression of T cell responses by arginase-induced L-arginine depletion in nonhealing leishmaniasis. PLoS Negl Trop Dis. 2009;3(7):e480. [DOI:10.1371/journal.pntd.0000480] [PMID] [PMCID]
78. Dubie T, Mohammed Y. Review on the Role of Host Immune Response in Protection and Immunopathogenesis during Cutaneous Leishmaniasis Infection. J Immunol Res. 2020;2020:2496713. [DOI:10.1155/2020/2496713] [PMID] [PMCID]
79. Aurora R, Creamer TP, Srinivasan R, Rose GD. Local interactions in protein folding: lessons from the α-helix. J Biol Chem. 1997;272(3):1413-6. [DOI:10.1074/jbc.272.3.1413] [PMID]
80. Argos P. An investigation of oligopeptides linking domains in protein tertiary structures and possible candidates for general gene fusion. J Mol Biol. 1990;211(4):943-58. [DOI:10.1016/0022-2836(90)90085-Z]
81. Huang Z, Zhang C, Xing X-H. Design and construction of chimeric linker library with controllable flexibilities for precision protein engineering. Meth Enzymol. 647: Elsevier; 2021. p. 23-49. [DOI:10.1016/bs.mie.2020.12.004] [PMID]
82. Rodrigues V, Cordeiro-da-Silva A, Laforge M, Silvestre R, Estaquier J. Regulation of immunity during visceral Leishmania infection. Parasites Vectors. 2016;9(1):118. [DOI:10.1186/s13071-016-1412-x] [PMID] [PMCID]
83. Baghbeheshti S, Hadadian S, Eidi A, Pishkar L, Rahimi H. Effect of flexible and rigid linkers on biological activity of recombinant tetramer variants of S3 antimicrobial peptide. Int J Pept Res Ther. 2021;27(1):457-62. [DOI:10.1007/s10989-020-10095-7]
84. Bai Y, Ann DK, Shen W-C. Recombinant granulocyte colony-stimulating factor-transferrin fusion protein as an oral myelopoietic agent. Proc Natl Acad Sci U S A. 2005;102(20): 7292-6. [DOI:10.1073/pnas.0500062102] [PMID] [PMCID]
85. Takamatsu N, Watanabe Y, Yanagi H, Meshi T, Shiba T, Okada Y. Production of enkephalin in tobacco protoplasts using tobacco mosaic virus RNA vector. FEBS letters. 1990;269(1):73-6. [DOI:10.1016/0014-5793(90)81121-4]

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2024 CC BY-NC 4.0 | Iranian Journal of Medical Microbiology

Designed & Developed by : Yektaweb | Publisher: Farname Inc