ArticlePDF Available

Mining Autoimmune-Disorder-Linked Molecular-Mimicry Candidates in Clostridioides difficile and Prospects of Mimic-Based Vaccine Design: An In Silico Approach

Authors:

Abstract and Figures

Molecular mimicry, a phenomenon in which microbial or environmental antigens resemble host antigens, has been proposed as a potential trigger for autoimmune responses. In this study, we employed a bioinformatics approach to investigate the role of molecular mimicry in Clostridioides difficile-caused infections and the induction of autoimmune disorders due to this phenomenon. Comparing proteomes of host and pathogen, we identified 23 proteins that exhibited significant sequence homology and were linked to autoimmune disorders. The disorders included rheumatoid arthritis, psoriasis, Alzheimer’s disease, etc., while infections included viral and bacterial infections like HIV, HCV, and tuberculosis. The structure of the homologous proteins was superposed, and RMSD was calculated to find the maximum deviation, while accounting for rigid and flexible regions. Two sequence mimics (antigenic, non-allergenic, and immunogenic) of ≥10 amino acids from these proteins were used to design a vaccine construct to explore the possibility of eliciting an immune response. Docking analysis of the top vaccine construct C2 showed favorable interactions with HLA and TLR-4 receptor, indicating potential efficacy. The B-cell and T-helper cell activity was also simulated, showing promising results for effective immunization against C. difficile infections. This study highlights the potential of C. difficile to trigger autoimmunity through molecular mimicry and vaccine design based on sequence mimics that trigger a defensive response.
Content may be subject to copyright.
Citation: Alshamrani, S.; Mashraqi,
M.M.; Alzamami, A.; Alturki, N.A.;
Almasoudi, H.H.; Alshahrani, M.A.;
Basharat, Z. Mining
Autoimmune-Disorder-Linked
Molecular-Mimicry Candidates in
Clostridioides difficile and Prospects of
Mimic-Based Vaccine Design: An In
Silico Approach. Microorganisms 2023,
11, 2300. https://doi.org/10.3390/
microorganisms11092300
Academic Editors: Amedeo Amedei
and Gwendolyn Barceló-Coblijn
Received: 1 July 2023
Revised: 7 September 2023
Accepted: 7 September 2023
Published: 12 September 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
microorganisms
Article
Mining Autoimmune-Disorder-Linked Molecular-Mimicry
Candidates in Clostridioides difficile and Prospects of
Mimic-Based Vaccine Design: An In Silico Approach
Saleh Alshamrani 1, Mutaib M. Mashraqi 1, * , Ahmad Alzamami 2, Norah A. Alturki 3,
Hassan H. Almasoudi 1, Mohammed Abdulrahman Alshahrani 1and Zarrin Basharat 4, *
1Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University,
Najran 61441, Saudi Arabia; saalshamrani@nu.edu.sa (S.A.); hhalmasoudi@nu.edu.sa (H.H.A.);
maalshahrani@nu.edu.sa (M.A.A.)
2Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University,
AlQuwayiyah 11961, Saudi Arabia; aalzamami@su.edu.sa
3Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University,
Riyadh 11433, Saudi Arabia; noalturki@ksu.edu.sa
4Alpha Genomics (Private) Limited, Islamabad 45710, Pakistan
*Correspondence: mmmashraqi@nu.edu.sa (M.M.M.); zarrin.iiui@gmail.com (Z.B.)
Abstract:
Molecular mimicry, a phenomenon in which microbial or environmental antigens resemble
host antigens, has been proposed as a potential trigger for autoimmune responses. In this study, we
employed a bioinformatics approach to investigate the role of molecular mimicry in Clostridioides
difficile-caused infections and the induction of autoimmune disorders due to this phenomenon.
Comparing proteomes of host and pathogen, we identified 23 proteins that exhibited significant
sequence homology and were linked to autoimmune disorders. The disorders included rheumatoid
arthritis, psoriasis, Alzheimer’s disease, etc., while infections included viral and bacterial infections
like HIV, HCV, and tuberculosis. The structure of the homologous proteins was superposed, and
RMSD was calculated to find the maximum deviation, while accounting for rigid and flexible regions.
Two sequence mimics (antigenic, non-allergenic, and immunogenic) of
10 amino acids from these
proteins were used to design a vaccine construct to explore the possibility of eliciting an immune
response. Docking analysis of the top vaccine construct C2 showed favorable interactions with
HLA and TLR-4 receptor, indicating potential efficacy. The B-cell and T-helper cell activity was also
simulated, showing promising results for effective immunization against C. difficile infections. This
study highlights the potential of C. difficile to trigger autoimmunity through molecular mimicry and
vaccine design based on sequence mimics that trigger a defensive response.
Keywords: Clostridioides difficile; molecular mimicry; autoimmunity; vaccine design; bioinformatics;
immune response; docking; pathogenesis
1. Introduction
Molecular mimicry refers to the phenomenon in which microbial or environmental
antigens share structural or sequence similarities with host antigens [
1
]. This similarity
can lead to a cross-reactive immune response, in which the immune system mistakenly
targets self-tissues, resulting in autoimmune disorders [
2
]. The onset of autoimmune
disorders due to molecular mimicry by pathogenic proteins or antigens presents an intrigu-
ing research area that investigates the possible connection between microbial infections
and the onset of autoimmune responses [
3
,
4
]. Understanding the mechanisms by which
molecular mimicry contributes to autoimmune disorders is crucial for developing tar-
geted therapies and preventive strategies [
5
7
]. Numerous studies have investigated the
molecular mimicry between human proteins associated with autoimmune disorders and
Microorganisms 2023,11, 2300. https://doi.org/10.3390/microorganisms11092300 https://www.mdpi.com/journal/microorganisms
Microorganisms 2023,11, 2300 2 of 25
pathogen-derived proteins [
8
13
]. These investigations have revealed several mechanisms
that contribute to the initiation and perpetuation of autoimmune responses [
14
16
]. Specific
pathogen proteins have been found to share sequence motifs with host proteins involved
in autoimmune disorders, enabling the activation of autoreactive T cells [
14
,
17
]. Strepto-
coccus pyogenes has been implicated in autoimmune diseases, such as rheumatic fever [
18
],
glomerulonephritis [
19
], and multiple sclerosis [
20
]. Epstein–Barr virus has been connected
with molecular-mimicry-mediated autoimmune disorders [
21
], like systemic lupus ery-
thematosus [
22
,
23
], hepatitis [
24
], and multiple sclerosis [
25
]. These pathogens possess
proteins that mimic self-antigens, leading to cross-reactivity with host tissues. Additionally,
molecular mimicry has been observed in viral infections, such as the hepatitis C virus, in
which viral proteins share sequence homology with host proteins involved in autoimmune
liver diseases [26].
Techniques that can be employed to study molecular-mimicking peptides include
phage display [
27
], and bioinformatics approaches include sequence alignment and molec-
ular modeling [
28
], etc. Sequence alignment algorithms can identify regions of similar-
ity or shared motifs between the two protein sequences [
29
]. Homology modeling and
comparative protein structure prediction can be used to analyze the three-dimensional
structures [
30
] of pathogen and host proteins. By comparing the structural features and
folding patterns, potential mimicking regions can be identified. Predictive algorithms, such
as NetMHC [
31
] and the Immune Epitope Database (IEDB) [
32
], can be utilized to identify
potential epitopes within pathogen proteins that resemble host epitopes associated with
autoimmune disorders [
33
]. To validate the cross-reactivity of pathogen and host proteins,
enzyme-linked immunosorbent assay (ELISA) assays [
34
], Western blotting [
35
], and flow
cytometry [
36
], etc., can be employed. These methods measure the binding of antibodies or
T cells to specific antigens and can confirm the presence of molecular mimicry. Apart from
these, animal models, such as transgenic mice expressing human proteins associated with
autoimmune disorders, can be used to study the effects of pathogen infections and eval-
uate the development of autoimmune responses [
37
,
38
]. Disease models, such as
in vitro
models of tissue inflammation can also be employed to investigate the consequences of
molecular mimicry [
8
,
39
]. By utilizing a combination of these techniques, researchers can
gain insights into the occurrence and mechanisms of molecular mimicry, contributing to
a better understanding of its role in autoimmune disorders and potentially guiding the
development of therapeutic interventions.
Bioinformatics is a swift approach to identifying and characterizing the molecular-
mimicry interactions between human proteins and pathogens [
40
42
]. Various bioinfor-
matics tools and databases are utilized to analyze protein sequences, identify shared motifs
or structural similarities, and predict antigenicity and immunogenicity [43]. Comparative
genomics and proteomics approaches are employed to identify pathogen proteins that
mimic host antigens associated with autoimmune disorders [
44
46
]. Previous studies
have utilized bioinformatics approaches to uncover potential molecular-mimicry mecha-
nisms between pathogens and host proteins by employing sequence alignment algorithms,
structural modeling, and epitope prediction tools to assess the extent of mimicry and
the potential immunological consequences [
15
,
41
,
47
]. Additionally, database-mining tech-
niques have been used to establish links between identified mimicry interactions and
autoimmune disorders [
9
,
48
50
]. Herein, we analyzed mimicry prediction and association
with autoimmune disorders in Clostridioides difficile using in silico methods. By integrating
bioinformatics analyses with experimental validation, this information contributes to our
understanding of the complex interactions between pathogens and the human immune sys-
tem, shedding light on the role of molecular mimicry in the development and progression
of autoimmune disorders.
Microorganisms 2023,11, 2300 3 of 25
2. Material and Methods
2.1. Homology Analysis
The entire set of proteins from the human and C. difficile S-0253 (reference strain
ASM1888508v1) samples was obtained from Uniprot (https://www.uniprot.org/proteomes/
UP000005640 (accessed on 31 May 2023)) and the NCBI database (GenBank accession:
CP076401.1; accessed 31 May 2023), respectively. To identify potential homologous proteins,
a local installation of BLAST was utilized, applying a threshold of
50% identity and
100-bit score to retain proteins for further analysis [16].
2.2. Mimic Region Identification
To identify potential regions involved in mimicry, the proteins were aligned to un-
cover regions of similarity with a minimum length of 10 amino acids [
16
]. RMSD was
employed as a scoring metric to assess the structural similarity of the peptides. To ob-
tain the 3D structures of these proteins, the state-of-the-art-predicted protein structures
from the AlphaFold database were utilized [
51
,
52
]. To compare and align the obtained
structures, both iPBA [
53
] and TM-align [
54
] algorithms were employed. These tools
are widely recognized tools in the field of structural biology, known for their accuracy
in comparing protein structures and determining alignment based on various structural
features. iPBA is a sequence-independent method that uses a fragment-based approach (for
capturing large protein fold changes) [
53
], while TM-align superimposes 3D coordinates
and aligns protein structures by dynamic programming method (for capturing small fold
changes) [
54
]. The superposed structures diagram was generated through the TM-align
module of the RCSB PDB structural alignment tool (https://www.rcsb.org/alignment
(accessed on 21 June 2023)).
2.3. Autoimmunity Elucidation
In order to identify homologous protein pathways associated with autoimmune
disorders or infection, relevant databases, such as pathDIP [
55
] and PHAROS (https:
//pharos.nih.gov/targets/ (accessed on 15 June 2023)) [
56
], were surveyed, along with a
thorough review of the literature. PHAROS provides preprocessed data from the Target
Central Resource Database (TCRD) on the input of the human gene name, Uniprot ID, etc.
Linked disorders can be manually checked for autoimmunity. For pathDIP, all databases
were selected with a minimum confidence level set to 0.99. The data type selected was
extended pathway associations. The protein interaction set considered for analysis included
both experimentally detected and computationally predicted protein–protein interactions
(PPIs) using the full IID dataset. pathDIP serves as a comprehensive reference for signaling
cascades across various species, consolidating key pathways sourced from major curated
pathway databases [
57
]. The associations in pathDIP are based on a combination of com-
putational predictions, experimentally confirmed interactions, orthology mapping, and
inference of physical protein interactions. This database provides a valuable resource for
exploring and understanding signaling pathways associated with autoimmune disorders
and infection. Apart from this, a literature search was conducted to identify infection or
autoimmune pathways linked with these homologs. A BLAST search (
90% homology) of
epitopes was also carried out against IEDB [
32
], and the relevant literature was identified
for the listed infection or autoimmune disorder in the database.
2.4. Mimic-Based Vaccine Design
Identified mimics were subjected to antigenic analysis using VaxiJen server [
58
]. Apart
from this, properties like allergenicity, toxicity, and other parameters useful in finalizing
peptides for vaccine design were studied. ProtParam was used for physicochemical pro-
filing [
59
], while AllerCatPro (https://allercatpro.bii.a-star.edu.sg/, accessed on 16 June
2023) and ToxinPred (https://webs.iiitd.edu.in/raghava/toxinpred/multi_submit.php,
accessed on 16 June 2023) were used for allergenicity and toxicity analysis, respectively.
IEDB was used for immunogenicity prediction [
32
]. Mimics were prioritized for vac-
Microorganisms 2023,11, 2300 4 of 25
cine design based on these parameters, the ability to induce immune response, and con-
servation across the strains. Conservations was determined by the ConSurf webserver
(https://consurf.tau.ac.il/consurf_index.php; accessed on 17 June 2023).
The vaccine construct was designed according to the previously described methodol-
ogy [
16
], using suitable linkers, adjuvants, and binders. They were subjected to another
round of evaluation according to properties like antigenicity, toxicity, allergenicity, etc.
The best antigenic and non-allergenic, non-toxic construct was tested for immune reaction
incitation and cloned in a pET-28(a)+ vector (available at https://www.snapgene.com/
plasmids/pet_and_duet_vectors_(novagen)/pET-28a(%2B); accessed on 18 June 2023) after
reverse translation and codon optimization through the JCat tool [
60
]. C-ImmSimm [
61
]
was used for the simulation of immune reaction. The parameters were as follows: Simu-
lation_volume = 10; Num_steps = 1000; HLA = A0101, A0102, B0702, B0704, DRB1_0101,
DRB1_0102; No_of_injections = 3; Time of injection (in days) = 1, 30); Adjuvant = 100.
The first two injections on day 1 and 30 were of the vaccine. At day 240, the proteins
phosphoribosylaminoimidazolecarboxamide formyltransferase and adenylosuccinate lyase
were injected to test the reaction.
2.5. Immune Receptor Binding Study
To elicit protection, the vaccine protein should bind the immune receptors with good
affinity [
62
,
63
]. To analyze this property, we constructed a 3D model of the vaccine construct
with SWISS-MODEL [
64
], AlphaFold [
51
], and I-TASSER [
65
]. The best model was selected
based on Ramachandran plot statistics from the assess module of SWISS-MODEL (https:
//swissmodel.expasy.org/assess (accessed on 20 June 2023)) and energy-minimized using
Molecular Operating Environment (MOE) v2016 software. It was then docked with immune
receptors of importance like TLR-4 receptor (PDB ID: 3FXI), HLA-A (PDB ID: 3OX8), and
HLA-B (PDB ID: 4JQX), using the ClusPro server [
66
]. ClusPro focuses on predicting the
overall shape and orientation of the protein–protein complex [
67
]. Prodigy [
68
] was used
to predict the thermodynamic properties and binding affinities of the obtained docked
complexes. An experimentally determined protein–protein interacting complex (PDB
ID:4GIQ) was employed as a control to compare predicted values. This comparison allowed
us to assess whether the binding scores were superior or inferior to the control, providing
valuable insights into the efficacy and specificity of our vaccine design approach.
3. Results
3.1. Homologous Sequence Identification
In total, 23 proteins were obtained with significant similarity between human and
C. difficile. F0F1 ATP synthase subunit beta had the highest number of peptide mimics
(n= 11), followed by F0F1 ATP synthase subunit alpha (n= 7) and heat-shock protein
DnaK (n= 6) (Table 1). These were superposed (Figure 1), and RMSD after structural
superposition varied between iPBA and TM-align prediction. A possible reason for this is
the algorithm difference, in which iPBA is tailored for flexible proteins or regions within
a protein. Thus, it gave overall lower RMSD values compared to TM-align. However,
both servers gave the lowest RMSD of 0.51 for the ATP-dependent Clp endopeptidase
proteolytic subunit ClpP. This suggests a high degree of structural similarity between the
human and bacterial homologs of this protein.
Microorganisms 2023,11, 2300 5 of 25
Table 1. RMSD of C. difficile homologs in humans. Molecular mimics with length 10 are also shown.
Serial
no. Name
UniProt ID
of Human
Homolog
NCBI Accession
of Bacterial
Homolog
Bacterial Protein
Structure
AlphaFold ID
No. of Similar
Peptides
(Length 10)
Molecular Mimic Region (Length 10)
Superposed
Protein RMSD
(iPBA)
TM-Align
RMSD
1Molecular chaperone DnaK P38646 QWS53804.1 Q182E8 6
GIDLGTTNSCVAV
RTTPSVVAFT
INEPTAAALAYG
LLLDVTPLSLGIET
RNTTIPTKKSQ
PQIEVTFDIDANGIV
1.13 2.0
2Translation elongation factor 4 Q8N442 QWS53810.1 Q182F4 3 DHGKSTLADRL
LNLIDTPGHVDF
VLAKCYGGDI 1.25 1.55
3Uracil-DNA glycosylase P13051 QWS53824.1 Q182G9 0 - 1.01 1.13
4Acetyl-CoA C-acetyltransferase Q9BWD1 QWS54012.1 Q18AR0 1 NASGINDGAA 0.70 0.75
5
3-oxoacid CoA-transferase subunit B
E9PDW2 QWS54014.1 Q183B1 0 - 1.00 1.74
6UDP-glucose 4-epimerase GalE Q14376 QWS54050.1 Q183E8 3 GGAGYIGSHT
VFSSSATVYG
DGTGVRDYIHV 0.79 0.79
7V-type proton ATPase subunit B P21281 QWS54278.1 Q184E3 2 YAEALREVSAA
THPIPDLTGYITEGQI 0.81 0.91
8
V-type ATP synthase catalytic unit A
P38606 QWS54279.1 Q184E7 2 MPVAAREASIYTGIT
MADSTSRWAEALRE 0.98 1.03
9Phosphopyruvate hydratase P09104 QWS54488.1 Q181T5 3 LDSRGNPTVEV
QEFMILPVGA
VGDEGGFAPN 0.87 1.39
10 ATP-dependent Clp endopeptidase
proteolytic subunit ClpP Q16740 QWS54701.1 Q180F0 1 VVEQTGRGER 0.51 0.51
11 ATP-dependent Clp endopeptidase
proteolytic subunit ClpP Q16740 QWS54730.1 Q180J6 0 - 0.56 0.56
12 F0F1 ATP synthase subunit beta P06576 QWS54823.1 Q184E3 11
TGIKVVDLLAPY
KGGKIGLFGGAGVGKTVLI
G VGERTREGNDLY
GQMNEPPGAR
DNIFRFTQAGSEVSALLGR
PSAVGYQPTLAT
TKKGSITSVQA
YVPADDLTDPAPATTF
LGIYPAVDPL
LQDIIAILGMDELS
RARKIQRFLSQ
2.11 2.73
Microorganisms 2023,11, 2300 6 of 25
Table 1. Cont.
Serial
no. Name
UniProt ID
of Human
Homolog
NCBI Accession
of Bacterial
Homolog
Bacterial Protein
Structure
AlphaFold ID
No. of Similar
Peptides
(Length 10)
Molecular Mimic Region (Length 10)
Superposed
Protein RMSD
(iPBA)
TM-Align
RMSD
13 F0F1 ATP synthase subunit alpha P25705 QWS54825.1 Q184E7 7
IKEGDIVKRTG
PIGRGQRELIIGDRQTGKTSI
CIYVAIGQKRST
YTIVVSATAS
YDDLSKQAVAYR
MSLLLRRPPGREAYPGDVFYLHSRLLERAAK
IETQAGDVSAYIPTNVISITDGQI
1.52 2.69
14 Elongation factor Tu P49411 QWS55098.1 Q18CE4 3 GTIGHVDHGKTTLTAAITK
DCPGHADYVKNMITG
DGPMPQTREH 0.98 1.12
15 Elongation factor Tu P49411 QWS55112.1 Q18CE4 3 GTIGHVDHGKTTLTAAITK
DCPGHADYVKNMITG
DGPMPQTREH 0.98 1.12
16 Methionine adenosyltransferase P31153 QWS55174.1 Q18CL7 4
EGHPDKICDQISD
RFVIGGPQGD
HGGGAFSGKD
TKVDRSAAYAAR
0.72 0.93
17 Chaperonin GroEL P10809 QWS55239.1 Q18CT5 3 AGDGTTTATVLA
VVAVKAPGFGD
DALNATRAAVEEGIV 1.22 3.61
18 Isocitrate/isopropylmalate
dehydrogenase family protein P50213 QWS55807.1 Q18A33 5
VTLIPGDGIGPE
VMPNLYGDILSDL
AGDGTTTATVLA
VVAVKAPGFGD
DALNATRAAVEEGIV
0.89 1.48
19 Phosphoribosylaminoimidazole-
carboxamide formyltransferase P31939 QWS55813.1 Q18A34 5
WQLVKELKEA
SFKHVSPAGAAVG
REVSDGIIAPGY
KYTQSNSVCYAK
GAGQQSRIHCTRLAG
0.73 1.23
20 Acyl-CoA dehydrogenase P16219 QWS55947.1 Q18AQ1 2 LIFEDCRIPK
ITEIYEGTSE 0.72 0.99
21 Acetyl-CoA C-acetyltransferase Q9BWD1 QWS55952.1 Q18AR0 2 NASGINDGAA 0.70 0.75
22 Fe-S cluster assembly scaffold
protein NifU Q9H1K1 QWS56138.1 Q18BE3 2 GCGSAIASSS 1.01 1.39
23 Adenylosuccinate lyase P30566 QWS56192.1 Q18BJ9 2 RGVKGTTGTQASFL
YKRNPMRSER 0.81 0.94
Microorganisms 2023,11, 2300 7 of 25
Microorganisms 2023, 11, x FOR PEER REVIEW 7 of 25
Figure 1. Superposed structures of the C. dicile and human homologous proteins. (A) Molecular
chaperone DnaK; (B)Translation elongation factor 4; (C) Uracil-DNA glycosylase; (D) Acetyl-CoA
C-acetyltransferase; (E) 3-oxoacid CoA-transferase subunit B; (F) UDP-glucose 4-epimerase GalE;
(G) V-type proton ATPase subunit B; (H) V-type ATP synthase catalytic unit A; (I) Phosphopyruvate
hydratase; (J) ATP-dependent Clp endopeptidase proteolytic subunit ClpP; (K) ATP-dependent Clp
endopeptidase proteolytic subunit ClpP; (L) F0F1 ATP synthase subunit beta. Due to space con-
straints, the rst 12 (Table 1) of the 23 proteins are shown here. Human homologs are shown in
brown and bacterial proteins are shown in blue.
3.2. Autoimmunity Prediction
PHAROS and PATHDIP database scan revealed several autoimmune diseases linked
with the human homologous sequences of C. dicile (Table 2). ATP-dependent Clp prote-
ase proteolytic subunit and elongation factor Tu had two copies, so homologs were re-
moved from database mining. For the rest of the homologs, the most commonly identied
infection was tuberculosis (for DnaK, V-type ATP synthase, and ClpP) and the most com-
monly identied autoimmune disease was rheumatoid arthritis (for chaperones DnaK
and GroEL, elongation factor tu, Translation elongation factor 4, 3-oxoacid CoA-transfer-
ase subunit B, V-type proton ATPase, phosphopyruvate hydratase, Phosphoribosylami-
noimidazolecarboxamide formyltransferase, and NifU).
Figure 1.
Superposed structures of the C. difficile and human homologous proteins. (
A
) Molecular
chaperone DnaK; (
B
) Translation elongation factor 4; (
C
) Uracil-DNA glycosylase; (
D
) Acetyl-CoA
C-acetyltransferase; (
E
) 3-oxoacid CoA-transferase subunit B; (
F
) UDP-glucose 4-epimerase GalE;
(
G
) V-type proton ATPase subunit B; (
H
) V-type ATP synthase catalytic unit A; (
I
) Phosphopyruvate
hydratase; (
J
) ATP-dependent Clp endopeptidase proteolytic subunit ClpP; (
K
) ATP-dependent
Clp endopeptidase proteolytic subunit ClpP; (
L
) F0F1 ATP synthase subunit beta. Due to space
constraints, the first 12 (Table 1) of the 23 proteins are shown here. Human homologs are shown in
brown and bacterial proteins are shown in blue.
3.2. Autoimmunity Prediction
PHAROS and PATHDIP database scan revealed several autoimmune diseases linked
with the human homologous sequences of C. difficile (Table 2). ATP-dependent Clp protease
proteolytic subunit and elongation factor Tu had two copies, so homologs were removed
from database mining. For the rest of the homologs, the most commonly identified infec-
tion was tuberculosis (for DnaK, V-type ATP synthase, and ClpP) and the most commonly
identified autoimmune disease was rheumatoid arthritis (for chaperones DnaK and GroEL,
elongation factor tu, Translation elongation factor 4, 3-oxoacid CoA-transferase subunit B,
V-type proton ATPase, phosphopyruvate hydratase, Phosphoribosylaminoimidazolecar-
boxamide formyltransferase, and NifU).
Microorganisms 2023,11, 2300 8 of 25
Table 2. Autoimmune pathways of the selected homologs.
Serial
no. Protein Homolog
PHAROS PATHDIP Literature
Autoimmunity
Pathway Infection Pathway Autoimmune
Pathway Infection Pathway Autoimmune Pathway Infection Pathway
1 P38646 Molecular chaperone DnaK Autoimmune
disease, Parkinson’s
disease
Perinatal necrotizing
enterocolitis, HIV,
Tuberculosis
Parkinson’s disease,
Huntington’s
disease, Diabetes
mellitus,
Alzheimer’s
Tuberculosis, HIV,
Papillomavirus,
E. coli,
Cytomegalovirus,
Staphylococcus sp.,
Legionellosis,
Chagas,
Leishmaniasis,
Measles
Guillain–Barrésyndrome [69],
Multiple sclerosis [70],
Vitiligo [71], Systemic lupus
erythematosus [72],
Ankylosing spondylitis [73],
Type I Diabetes mellitus [74],
Rheumatoid arthritis [75]
Trypanosoma
cruzi [76],
Mycobacterium
leprae [77]
2 Q8N442 Translation elongation factor 4 - - Tuberculosis,
Rheumatoid arthritis
- -
3 P13051 Uracil-DNA glycosylase ---HIV, Viral
carcinogenesis - -
4 Q9BWD1 Acetyl-CoA C-acetyltransferase ---HBV, Viral
carcinogenesis Systemic lupus
erythematosus [78]HCV [79]
5 E9PDW2 3-oxoacid CoA-transferase subunit B Crohn’s disease ---Rheumatoid arthritis [80]-
6 Q14376 UDP-glucose 4-epimerase GalE Psoriasis, Interstitial
cystitis Tinea corporis,
Tinea pedis - - Type I Diabetes mellitus [81]Hemophilus
influenzae [82]
7 P21281 V-type proton ATPase subunit B IgA
glomerulonephritis -Huntington’s
disease, Rheumatoid
arthritis
Helicobacter pylori
infection, HPV,
Tuberculosis, Viral
carcinogenesis, Vibrio
chloerae, HIV
-Tuberculosis [83],
SARS-CoV-2 [84]
8 P38606 V-type ATP synthase catalytic unit A Psoriasis, Myopathy -
Alzheimer’s disease,
Huntington’s
disease, Parkinson’s
disease, Rheumatoid
arthritis,
HPV, H. pylori,
Tuberculosis, Vibrio
cholerae, Viral
carcinogenesis, HIV
Thyroid eye disease [85]
Influenza H1N1 [86],
Salmonellosis [87],
Rabies virus [88],
SARS-CoV-2 [89],
Tuberculosis [90]
9 P09104 Phosphopyruvate hydratase Psoriasis - - - Autoimmune
encephalomyelitis [91],
Rheumatoid arthritis [92]
Cytomegalovirus
[93]
10 Q16740 ATP-dependent Clp endopeptidase
proteolytic subunit ClpP Psoriasis -
Alzheimer’s disease,
Huntington’s
disease, Parkinson’s
disease
- - Tuberculosis [94]
Microorganisms 2023,11, 2300 9 of 25
Table 2. Cont.
Serial
no. Protein Homolog
PHAROS PATHDIP Literature
Autoimmunity
Pathway Infection Pathway Autoimmune
Pathway Infection Pathway Autoimmune Pathway Infection Pathway
11 P06576 F0F1 ATP synthase subunit beta - -
Alzheimer’s,
Huntington’s,
Parkinson’s,
Non-alcoholic
fatty-acid liver
diseases
Epstein–Barr virus
infection, HBV, HCV,
HPV, Measles,
Legionellosis, E. coli
Autoimmune myocarditis [95]
MERS
coronavirus [96],
Echinococcus
granulosus [97]
12 P25705 F0F1 ATP synthase subunit alpha Alzheimer’s disease ---Sjogren’s syndrome [98],
Crohn’s disease [98],
Ankolysing spondolytis [73]-
13 P49411 Elongation factor Tu - - Huntington’s,
Parkinson’s
HCV, HBV,
Legionellosis,
E. coli,V. cholerae
Sjogren’s syndrome [99],
Crohn’s disease [98],
Ankolysing spondylitis [73]
Streptococcus
pneumoniae [100],
bacteria like Bacillus
anthracis,Francisella
talurensis,
Staphylococcus sp.,
E. coli,H. pylori,
etc. [101]
14 P31153 Methionine adenosyltransferase
Type 2 diabetes
mellitus,
demyelinating
diseases, MODY,
Psoriasis, fatty liver,
or non-alcoholic
steatohepatitis
---Rheumatoid arthritis [102],
Uveitis [103]
Herpes simplex
type 1 [104],
Poxvirus [105], West
Nile virus [106]
15 P10809 Chaperonin GroEL Allergic rhinitis Tuberculosis, HIV - -
Type I Diabetes [103], Juvenile
chronic arthritis [107],
Atherosclerosis [108], Crohn’s
disease [109], Rheumatoid
arthritis [110], Systemic lupus
erythematosus [111], Sjogren
syndrome [112], Hashimoto
thyroiditis [113,114], and
myasthenia gravis [115],
Autism [116]
H. pylori [117],
P. aeruginosa and
S. aureus [118]
16 P50213 Isocitrate dehydrogenase [NAD]
subunit alpha Psoriasis - Huntington’s
disease, Parkinson’s
disease
Epstein–Barr virus,
HCV, Legionellosis Atherosclerosis [119],
Type I diabetes [120]H. pylori [117]
Microorganisms 2023,11, 2300 10 of 25
Table 2. Cont.
Serial
no. Protein Homolog
PHAROS PATHDIP Literature
Autoimmunity
Pathway Infection Pathway Autoimmune
Pathway Infection Pathway Autoimmune Pathway Infection Pathway
17 P31939 Phosphoribosylaminoimidaz-
olecarboxamide formyltransferase
Rheumatoid arthritis,
Psoriatic arthritis,
Erythrodermic
psoriasis, Pustular
psoriasis, Plaque
psoriasis, Diabetes
mellitus, Juvenile
idiopathic arthritis
- - - - C. neoformans [121]
18 P16219 Acyl-CoA dehydrogenase Allergic rhinitis,
Ulcerative colitis,
Crohn’s disease - - - - -
19 Q9BWD1 Acetyl-CoA C-acetyltransferase - - Parkinson’s disease HBV, Viral
carcinogenesis - -
20 Q9H1K1 Fe-S cluster assembly scaffold
protein NifU - - Parkinson’s disease Influenza, HIV -Human respiratory
syncytial virus [122],
SARS-CoV-1 [96]
21 P30566 Adenylosuccinate lyase Psoriasis - - - - Schistosomiasis [123],
Chlamydia sp. [124]
Microorganisms 2023,11, 2300 11 of 25
3.3. Sequence Mimics and Vaccine Design
Out of 68 mimics
10 aminoacids in length, 31 were antigenic (Table 3), with four being
allergenic. Among the 31 antigenic sequences, 14 mimics were identified as IL-4 inducers
(RTTPSVVAFT, DHGKSTLADRL, GGAGYIGSHT, DGTGVRDYIHV, LGIYPAVDPL, IKEGDI-
VKRTG, CIYVAIGQKRST, IETQAGDVSAYIPTNVISITDGQI, EGHPDKICDQISD, TKVDR-
SAAYAAR, GAGQQSRIHCTRLAG, GCGSAIASSS, RGVKGTTGTQASFL, YKRNPMRSER).
Only three peptides (PQIEVTFDIDANGIV, CIYVAIGQKRST, GAGQQSRIHCTRLAG), be-
longing to DnaK, F0F1 ATP synthase subunit alpha, and Adenylosuccinate lyase, were pre-
dicted as non-inducers of IL-6, while all the rest were inducers. Mimics LLLDVTPLSLGIET,
DGTGVRDYIHV, VGERTREGNDLY, EGHPDKICDQISD, and RGVKGTTGTQASFL, be-
longing to DnaK, GalE, F0F1 ATP synthase subunit beta, methionine adenosyltransferase,
and adenylosuccinate lyase, respectively, were predicted as inducers of IL-10.
Table 3. Immunogenicity, allergenicity, and other properties of the antigenic mimics.
Serial
no.
Antigenic
Score Sequence Length
Immuno-
genicity
Score
SVM Score
for Toxicity Hydroph-
obicity
Hydropath-
icity
Hydroph-
ilicity Charge Mol Wt Allergenicity
1. 1.09 GIDLGTTNSCVAV 15 0.65 0.07 0.85 0.42 1 1249.59 0.07 No
2. 0.58 RTTPSVVAFT 12 0.35 0.07 0.4 0.39 1 1078.36 0.07 No
3. 0.89 LLLDVTPLSLGIET 12 0.31 0.15 1.18 0.49 2 1483.99 0.15 yes
4. 1.39 PQIEVTFDIDANGIV 11 0.29 0.04 0.42 0.16 3 1631.04 0.04 yes
5. 0.80 DHGKSTLADRL 12 0.26 0.33 1.01 0.66 0.5 1212.48 0.33 No
6. 1.22 NASGINDGAA 12 0.26 0.05 0.22 0.04 1 889.02 0.05 No
7. 0.65 GGAGYIGSHT 24 0.21 0.08 0.13 0.52 0.5 919.11 0.08 No
8. 1.47 DGTGVRDYIHV 10 0.18 0.15 0.42 0.09 0.5 1231.49 0.15 No
9. 1.20 LDSRGNPTVEV 10 0.18 0.23 0.57 0.39 1 1186.44 0.23 yes
10. 1.48 VVEQTGRGER 11 0.14 0.42 1.26 0.88 0 1130.37 0.42 No
11. 0.54 TGIKVVDLLAPY 10 0.12 0.1 0.91 0.47 0 1288.73 0.1 No
12. 1.25 VGERTREGNDLY 11 0.12 0.41 1.48 0.77 1 1408.66 0.41 No
13. 1.03 PSAVGYQPTLAT 10 0.11 0.03 0.08 0.58 0 1204.51 0.03 No
14. 1.04 TKKGSITSVQA 14 0.11 0.19 0.38 0.2 2 1119.44 0.19 no
15. 0.54 LGIYPAVDPL 10 0.09 0.19 0.97 0.67 1 1057.4 0.19 yes
16. 0.65 IKEGDIVKRTG 10 0.06 0.29 0.69 0.86 1 1215.58 0.29 No
17. 0.70 CIYVAIGQKRST 12 0.05 0.13 0.2 0.23 2 1338.76 0.13 No
18. 0.53 YTIVVSATAS 10 0.02 0.14 1.22 0.83 0 1011.27 0.14 No
19. 0.84 IETQAGDVSAYIP
TNVISITDGQI 12 0.01 0.02 0.25 0.26 3 2506.13 0.02 No
20. 0.56 DGPMPQTREH 13 0.05 0.41 2.06 0.7 0.5 1167.4 0.41 No
21. 1.41 EGHPDKICDQISD 10 0.05 0.28 1.22 0.8 2.5 1456.73 0.28 No
22. 1.22 HGGGAFSGKD 12 0.06 0.1 0.84 0.28 0.5 932.1 0.1 No
23. 0.57 TKVDRSAAYAAR 15 0.12 0.35 0.65 0.51 2 1308.6 0.35 No
24. 1.75 AGDGTTTATVLA 12 0.13 0.06 0.52 0.28 1 1077.32 0.06 No
25. 1.07 VTLIPGDGIGPE 10 0.16 0.11 0.41 0.11 2 1167.51 0.11 No
26. 1.76 AGDGTTTATVLA 14 0.17 0.06 0.52 0.28 1 1077.32 0.06 No
27. 1.62 GAGQQSRIHCTRLAG 13 0.18 0.23 0.5 0.01 2.5 1554.97 0.23 No
28. 1.22 NASGINDGAA 10 0.19 0.05 0.22 0.04 1 889.02 0.05 No
29. 1.42 GCGSAIASSS 10 0.20 0.06 0.66 0.26 0 839.01 0.06 No
30. 1.65 RGVKGTTGTQASFL 11 0.22 0.14 0.24 0.07 2 1422.81 0.14 No
31. 0.55 YKRNPMRSER 11 0.25 0.77 2.62 1.19 3 1336.66 0.77 No
Two sequence mimics (GAGQQSRIHCTRLAG and RGVKGTTGTQASFL) belonging
to the phosphoribosylaminoimidazolecarboxamide formyltransferase and adenylosucci-
nate lyase proteins, respectively, were selected for vaccine design based on antigenicity and
other values. RGVKGTTGTQASFL was predicted as an inducer of IL-4, IL-6, and IL-10,
while GAGQQSRIHCTRLAG was predicted as only an IL-4 inducer. Evolutionary analysis
revealed majority of residues of both sequence mimics are highly conserved (Figure 2).
Microorganisms 2023,11, 2300 12 of 25
Microorganisms 2023, 11, x FOR PEER REVIEW 12 of 25
(A)
(B)
Figure 2. Conservation of sequence mimics from (A) phosphoribosylaminoimidazolecarboxamide
formyltransferase and (B) adenylosuccinate lyase used for vaccine design underlined by red (and
star symbol). Yellow color indicates insucient data for conservation inference.
Mimic-based vaccine design is an innovative approach that utilizes synthetic pep-
tides or proteins to mimic specic antigens of pathogens [16]. By presenting these mimics
to the immune system, it can generate targeted immune responses against the actual path-
ogen. Here, in total, nine constructs were made, and the non-allergenic ones were retained
for analysis (Supplementary Table S1). Among these, a stable, highly antigenic one, i.e.,
construct C2 was chosen for further downstream processing. It was reverse-translated and
cloned in a pET-28(a)+ vector (Figure 3).
Figure 2.
Conservation of sequence mimics from (
A
) phosphoribosylaminoimidazolecarboxamide
formyltransferase and (
B
) adenylosuccinate lyase used for vaccine design underlined by red (and
star symbol). Yellow color indicates insufficient data for conservation inference.
Mimic-based vaccine design is an innovative approach that utilizes synthetic peptides
or proteins to mimic specific antigens of pathogens [
16
]. By presenting these mimics to the
immune system, it can generate targeted immune responses against the actual pathogen.
Here, in total, nine constructs were made, and the non-allergenic ones were retained
for analysis (Supplementary Table S1). Among these, a stable, highly antigenic one, i.e.,
construct C2 was chosen for further downstream processing. It was reverse-translated and
cloned in a pET-28(a)+ vector (Figure 3).
3.4. Immune Response Simulation
Immune response simulation analysis utilized Parker’s propensity scale to predict
potential epitopes within the vaccine sequence [
125
], which may be recognized by the
immune system, particularly by T cells. Six B-cell epitopes (EQIG, STRGRKCCRRKKEA,
AGGGSRGVKGTTGT, AGGGSGAGQ, GGSHEY, AGGGS) were identified using the Parker
propensity scale. For MHC-I, no binding epitope was present for the A0101 and B0702
allele, while two binding epitopes (IINTLQKYY and AGGGGSHEY) were identified for the
A0102 allele and one (RVRGGRCAV) was identified for B0704. For MHC-II binding, seven
epitopes were predicted for DRB1_0101 (YCRVRGGRC, FVAAWTLKA, WTLKAAAGG,
LKAAAGGGS, VKGTTGTQA, FLGGGSAKF, LERAGAKFV) and two were predicted for
DRB1_0102 (LKAAAGGGS and LERAGAKFV).
Microorganisms 2023,11, 2300 13 of 25
Microorganisms 2023, 11, x FOR PEER REVIEW 13 of 25
Figure 3. The 5083 bp cloned vector of the vaccine construct.
3.4. Immune Response Simulation
Immune response simulation analysis utilized Parkers propensity scale to predict
potential epitopes within the vaccine sequence [125], which may be recognized by the im-
mune system, particularly by T cells. Six B-cell epitopes (EQIG, STRGRKCCRRKKEA,
AGGGSRGVKGTTGT, AGGGSGAGQ, GGSHEY, AGGGS) were identied using the Par-
ker propensity scale. For MHC-I, no binding epitope was present for the A0101 and B0702
allele, while two binding epitopes (IINTLQKYY and AGGGGSHEY) were identied for
the A0102 allele and one (RVRGGRCAV) was identied for B0704. For MHC-II binding,
seven epitopes were predicted for DRB1_0101 (YCRVRGGRC, FVAAWTLKA,
WTLKAAAGG, LKAAAGGGS, VKGTTGTQA, FLGGGSAKF, LERAGAKFV) and two
were predicted for DRB1_0102 (LKAAAGGGS and LERAGAKFV).
The IgM + IgG population was 140,000 cells/mm3, and only a slight dierence was
observed after the second injection (Figure 4A). This suggests that the primary immune
response, characterized by the production of IgM antibodies followed by IgG antibodies
[126,127], was already established after the rst injection. The second injection did not
result in a signicant increase in the overall IgM + IgG population. IgG1 + IgG2 count was
below 80,000 cells/mm3 but increased to more than 90,000 cells/mm3 after the second in-
jection. This indicates that the secondary immune response, mediated by IgG antibodies
[128], was robustly triggered by the second injection. The overall immune cell counts plat-
eaued after around 200 days, indicating stabilization of the immune response. The B-cell
population remained active after the vaccine injection, up to 100 cells/mm3 (Figure 4B).
Their sustained presence suggests ongoing immune surveillance and the potential for
long-term immune memory [129]. T-helper (TH) cells increased after each vaccine injec-
tion and remained active with a count of 4000 cells/mm3 even after 300 days (Figure 4C).
Figure 3. The 5083 bp cloned vector of the vaccine construct.
The IgM + IgG population was 140,000 cells/mm
3
, and only a slight difference
was observed after the second injection (Figure 4A). This suggests that the primary
immune response, characterized by the production of IgM antibodies followed by IgG
antibodies [126,127]
, was already established after the first injection. The second injection
did not result in a significant increase in the overall IgM + IgG population.
IgG1 + IgG2
count was below 80,000 cells/mm
3
but increased to more than 90,000 cells/mm
3
after the
second injection. This indicates that the secondary immune response, mediated by IgG
antibodies [
128
], was robustly triggered by the second injection. The overall immune cell
counts plateaued after around 200 days, indicating stabilization of the immune response.
The B-cell population remained active after the vaccine injection, up to 100 cells/mm
3
(Figure 4B). Their sustained presence suggests ongoing immune surveillance and the
potential for long-term immune memory [
129
]. T-helper (TH) cells increased after each
vaccine injection and remained active with a count of 4000 cells/mm3even after 300 days
(
Figure 4C
). TH cells play a crucial role in coordinating the immune response by facilitating
communication between various immune cells, and their persistent presence indicates their
continued involvement in supporting and regulating the immune response [
130
,
131
]. The
count of cytotoxic T (TC) non-memory cells fluctuated, possibly indicating their active
participation in eliminating target cells (Figure 4D). In contrast, TC memory cells remained
consistently higher than 100 cells/mm
3
, suggesting the establishment of immunological
memory. Memory cells enable a rapid and specific response upon re-exposure to the anti-
gen, contributing to long-term immunity [
132
]. Natural killer (NK) cells have a role in
innate immune defense [
133
], and their population remained at more than 300 cells/mm
3
for the whole period (Figure 4E). No significant changes were observed in response to
the stress of bacterial proteins. This suggests that the immune system reached a state of
equilibrium and was no longer strongly influenced by the presence of bacterial proteins.
Microorganisms 2023,11, 2300 14 of 25
Microorganisms 2023, 11, x FOR PEER REVIEW 14 of 25
TH cells play a crucial role in coordinating the immune response by facilitating commu-
nication between various immune cells, and their persistent presence indicates their con-
tinued involvement in supporting and regulating the immune response [130,131]. The
count of cytotoxic T (TC) non-memory cells uctuated, possibly indicating their active
participation in eliminating target cells (Figure 4D). In contrast, TC memory cells re-
mained consistently higher than 100 cells/mm3, suggesting the establishment of immuno-
logical memory. Memory cells enable a rapid and specic response upon re-exposure to
the antigen, contributing to long-term immunity [132]. Natural killer (NK) cells have a
role in innate immune defense [133], and their population remained at more than 300
cells/mm3 for the whole period (Figure 4E). No signicant changes were observed in re-
sponse to the stress of bacterial proteins. This suggests that the immune system reached a
state of equilibrium and was no longer strongly inuenced by the presence of bacterial
proteins.
Figure 4. Immune system cells released after the C2 vaccine and C. dicile protein (phosphoribosyl-
aminoimidazolecarboxamide formyltransferase and adenylosuccinate lyase) stress, including (A)
immunoglobulins, (B) B cells, (C) TH cells, (D) TC cells, and (E) NK cells.
Figure 4.
Immune system cells released after the C2 vaccine and C. difficile protein (phosphoribo-
sylaminoimidazolecarboxamide formyltransferase and adenylosuccinate lyase) stress, including
(A) immunoglobulins, (B) B cells, (C) TH cells, (D) TC cells, and (E) NK cells.
3.5. Vaccine Interaction
The 3D structure of the vaccine construct was modeled using three tools, in which
SWISS-MODEL achieved the highest percentage (89.60%), indicating a larger portion
of residues in favorable conformation compared to I-TASSER (44.32%) and AlphaFold
(62.75%) (Supplementary Table S2). The QMEANDisCo Global score was additionally used
to assess the global quality of the protein structure, with a lower score suggesting better
overall quality. This metric is used to estimate the quality of a protein tertiary structure by
taking distance constraints into account [
134
]. Again, SWISS-MODEL achieved a score of
0.64 ±0.07, followed by AlphaFold (0.35 ±0.07) and I-TASSER (0.32 ±0.07).
The best-modeled structure by SWISS-MODEL (Supplementary Figure S1A) was used
to map interactions with HLA and TLR-4 receptor (Figure 5). Docking revealed that HLA-A
and HLA-B complexes had relatively lower binding scores compared to the TLR-4 complex,
implying stronger binding affinities between the vaccine construct and HLA receptors
(Supplementary Table S3). The PRODIGY server [
68
] was used to map thermodynamic
Microorganisms 2023,11, 2300 15 of 25
changes in these complexes, where
G (kcal mol
1
) represents the change in free energy
associated with the formation of the protein–protein complex, while K
d
(M) provided the
equilibrium dissociation constant at 25
C.
G is studied to measure the stability of the
complex, while K
d
is studied to measure the binding affinity, with more negative values
suggesting a stronger interaction [
135
,
136
].
G and K
d
are better predictors of binding
than docking score [
137
] and were, therefore, employed for validation. HLA-B and TLR-4
had a highly negative
G value, indicating stable and stronger interaction compared to
the control. TLR-4 indicated the lowest K
d
value, suggesting a strong binding affinity
and a favorable binding interaction in comparison with the control. This suggests that
the interactions between the vaccine construct and TLR-4 receptor are likely to be more
favorable and specific. This also shows that the ClusPro modeling method performed well
in predicting the binding of the HLA and TLR-4 complex with vaccine construct and is a
reliable approach to determine interactions.
Microorganisms 2023, 11, x FOR PEER REVIEW 15 of 25
3.5. Vaccine Interaction
The 3D structure of the vaccine construct was modeled using three tools, in which
SWISS-MODEL achieved the highest percentage (89.60%), indicating a larger portion of
residues in favorable conformation compared to I-TASSER (44.32%) and AlphaFold
(62.75%) (Supplementary Table S2). The QMEANDisCo Global score was additionally
used to assess the global quality of the protein structure, with a lower score suggesting
beer overall quality. This metric is used to estimate the quality of a protein tertiary struc-
ture by taking distance constraints into account [134]. Again, SWISS-MODEL achieved a
score of 0.64 ± 0.07, followed by AlphaFold (0.35 ± 0.07) and I-TASSER (0.32 ± 0.07).
The best-modeled structure by SWISS-MODEL (Supplementary Figure S1A) was
used to map interactions with HLA and TLR-4 receptor (Figure 5). Docking revealed that
HLA-A and HLA-B complexes had relatively lower binding scores compared to the TLR-
4 complex, implying stronger binding anities between the vaccine construct and HLA
receptors (Supplementary Table S3). The PRODIGY server [68] was used to map thermo-
dynamic changes in these complexes, where ΔG (kcal mol1) represents the change in free
energy associated with the formation of the protein–protein complex, while Kd (M) pro-
vided the equilibrium dissociation constant at 25 . ΔG is studied to measure the stability
of the complex, while Kd is studied to measure the binding anity, with more negative
values suggesting a stronger interaction [135,136]. ΔG and Kd are beer predictors of bind-
ing than docking score [137] and were, therefore, employed for validation. HLA-B and
TLR-4 had a highly negative ΔG value, indicating stable and stronger interaction com-
pared to the control. TLR-4 indicated the lowest Kd value, suggesting a strong binding
anity and a favorable binding interaction in comparison with the control. This suggests
that the interactions between the vaccine construct and TLR-4 receptor are likely to be
more favorable and specic. This also shows that the ClusPro modeling method per-
formed well in predicting the binding of the HLA and TLR-4 complex with vaccine con-
struct and is a reliable approach to determine interactions.
Figure 5. Vaccine construct (shown in green) interaction with (A) HLA-A, (B) HLA-B, and (C) TLR-
4. Receptors are shown in cyan. (D) Control Tumor necrosis factor ligand superfamily member 11
(RANK-L) and 11A (RANK) from Mus musculus.
Figure 5.
Vaccine construct (shown in green) interaction with (
A
) HLA-A, (
B
) HLA-B, and (
C
) TLR-4.
Receptors are shown in cyan. (
D
) Control Tumor necrosis factor ligand superfamily member 11
(RANK-L) and 11A (RANK) from Mus musculus.
4. Discussion
C. difficile can cause infections, primarily in the colon or large intestine [
138
,
139
].
Infection usually occurs in the immunocompromised [
140
] and in people who have received
antibiotic therapy, when the natural balance of bacteria in the colon is disrupted [
138
].
This allows the bacterium to multiply and produce toxins that cause inflammation and
damage to the intestinal lining. Molecular mimicry allows C. difficile toxin A to bind
glycosphingolipids [
141
]. Mindur et al. have reported cross-reactive epitopes of myelin
basic protein in the surface layer protein of a sub-species of C. difficile [
142
]. Peptide
EQSLITVEGDKASM from the toxin B protein of the species has also been implicated in an
autoimmune disease, namely primary biliary cirrhosis. Alam et al. have reported a collagen
triple-helix repeat family protein in C. difficile as a mimic of the type II collagen protein of
humans [
143
]. The protein is implicated in reactive arthritis, septic arthritis, and rheumatic
symptoms. However, the sequence identity was less than 45%. This is why this protein
was missed by our analysis, as we followed stringent criteria of identity value 50%.
The molecular mimics at the whole proteome scale for C. difficile and their involvement
in autoimmune disorders have not yet been mapped. A bioinformatics-based approach
is a useful method to exploit the publicly available data for this purpose. Mapping the
Microorganisms 2023,11, 2300 16 of 25
molecular-mimicry mechanism employed by C. difficile can provide insights into the viru-
lence and pathogenesis, as well as offer potential targets for the development of therapeutic
interventions, such as vaccines or drugs that can disrupt the interaction between the bacte-
rial mimics and the host receptors, thereby preventing or reducing the severity of C. difficile
infections. For this purpose, we obtained the proteome of the reference strain of C. difficile
S-0253 (n= 3732 proteins). Among these, only 23 proteins were homologous to humans,
having
50% sequence identity. The structural superposition of these proteins revealed
several regions with organizational and fold similarity (Table 1). The ATP-dependent Clp
endopeptidase proteolytic subunit ClpP exhibited the lowest RMSD value, indicating a
high degree of structural similarity between the human and bacterial homologs. It plays
a crucial role in maintaining protein homeostasis in conjunction with chaperones by de-
grading misfolded or damaged proteins. The peptide sequence QIERDTERDRFLSAPEAV
of E. coli ClpP has been previously implicated in autoimmune biliary liver cirrhosis [
144
].
The highest number of peptide mimics were observed in energy-generating F0F1 ATP
synthase and heat-shock protein DnaK. Zhang et al. have reported increased activity of
ATP synthase in the autoimmune neuromyelitis optica spectrum disorder [
145
]. DnaK
has previously been implicated in molecular mimicry of other pathogenic bacteria like
Streptococcus pneumoniae [
16
] and Salmonella typhi [
15
]. DnaK and other molecular chaper-
ones like GroEL have been implicated in multiple autoimmune disorders [
116
,
146
151
].
DnaK has been associated with autoimmune atrophic gastritis caused by H. pylori [
152
],
while
Qeshmi et al.
have reported its presence in multiple sclerosis as well [
153
]. Overall,
rheumatoid arthritis, Alzheimer’s disease, psoriasis, Huntington’s disease, and Parkinson’s
disease emerged as the primary autoimmune disease associated with multiple homologs.
Among infectious disease mapping, tuberculosis was the most common infection
linked to the homologous proteins of C. difficile, suggesting a potential role of these proteins
in the immune response against mycobacterial infections. A varying fraction of C. difficile
infection in tuberculosis patients has been reported previously, ranging from ~3 cases
per 1000 adults in Korea [
154
] to ~70 cases per 1000 individuals in South Africa [
155
].
Obuch-Woszczaty´nski reported C. difficile-mediated diarrhea in tuberculosis patients when
rifampicin was used as part of their treatment regimen [
156
]. Rifampicin can contribute to
the development of resistance against this antibiotic in C. difficile, which in turn poses a risk
to the effectiveness of tuberculosis treatment. The rate of C. difficile infection in tuberculosis
patients tends to be higher in aged people compared to younger adults [
157
]. This bacterium
has also been identified as one of the predominant pathogens causing diarrheal illness
in HIV-seropositive individuals, with two times higher prevalence compared to HIV-
seronegative people [158].
Mimic-based vaccine design is an innovative approach that utilizes synthetic pep-
tides or proteins to mimic specific antigens of pathogens [
16
]. By presenting these mimics
to the immune system, targeted immune responses against the actual pathogen can be
generated [159,160]
. Hence, a stable and highly antigenic vaccine construct was designed
using two peptide mimics identified in this study. It was cloned into a pET-28(a)+ vector
and immune response was assessed using in silico simulations. The primary immune re-
sponse, characterized by IgM production followed by IgG production, was established after
the first vaccine injection. The secondary immune response, mediated by IgG antibodies,
was robustly triggered by the second injection. The immune cell counts plateaued after
approximately 200 days, indicating stabilization of the immune response. B cells remained
active, suggesting ongoing immune surveillance and potential long-term immune memory.
TH cells increased after each vaccine injection and remained active even after 300 days, indi-
cating their continued involvement in supporting and regulating the immune response. TC
cells showed fluctuating counts, possibly indicating their active participation in eliminating
target cells, while TC memory cells remained consistently higher, indicating the establish-
ment of immunological memory. NK cells, involved in innate immune defense, maintained
a stable population throughout the study. Hence, a dynamic and robust immune response
occurred following the vaccine injections. The presence of specific antibody populations
Microorganisms 2023,11, 2300 17 of 25
sustained B-cell activity, and a stable count of T cells, NK cells, and EP cells indicated an
effective immune response against the target antigen. The establishment of immunological
memory and the plateauing of immune cell counts suggests a stable and functional immune
system capable of long-term protection. No significant changes were observed in response
to bacterial protein stress, suggesting that the immune system reached an equilibrium state
and was no longer strongly influenced by the presence of bacterial proteins. However,
computational predictions are not without limitations, and although mimic-based vaccine
design and immune response simulation helps accelerate the vaccine-development process,
providing insights into immune responses and generating hypotheses for further exper-
imental investigations, it also has limitations. The foremost limitation is their accuracy
and adverse response mapping due to inadequate input of variables and complexities of
the immune system [
16
]. The local tissue microenvironment and factors such as blood
flow, physical barriers, and cellular interactions can influence immune responses but may
be overlooked or simplified in simulations. Additionally, the pathogens tend to mutate,
and they may not be workable in the real-world scenario due to the altered genetics of the
microbe. To overcome this, we have tried to focus on conserved epitopes of the antigenic
proteins, but the immune evasion mechanism may be altered with time and the epitope may
fail to generate an immune response. Thus, the in silico vaccine design is a valuable tool
for narrowing down potential candidates and reducing research costs and timelines, but it
is just the initial step in the vaccine-development process. Real laboratory-based testing is
essential to validate and refine these designs, ensuring that the vaccine candidates are safe
and effective in a real-world setting. Moreover, the in silico outcomes may not necessarily
mirror those in a real laboratory as they typically do not consider the environmental factors
the laboratory experiments take into account and can be overly optimistic or pessimistic
predictions. Biological systems can also have unexpected interactions and feedback loops
that are difficult to predict computationally. These interactions may only become apparent
through real-world experimentation. The limitations of in silico modeling highlight the
need for a comprehensive and rigorous approach to vaccine development that combines
computational methods with empirical testing.
In summary, using bacterial peptides as structural templates for vaccine design is
a valid approach, but the risk of triggering an autoimmune reaction prevails [
43
]. It is
crucial to assess the risk associated with the potential induction of autoimmune response
after administering the vaccine. It is also important to acknowledge that factors such as
prior exposure to antigens and the presence of known autoantibodies, genetic predisposi-
tion of individuals, and other variables may contribute to the initiation of cross-reactive
responses [161]
. However, the immune system is normally fortified with multiple lay-
ers of protective mechanisms, which work in concordance to prevent the occurrence of
autoimmunity [162]
in response to vaccines. The mammalian immune system has also
evolved an intricate repertoire of mechanisms to discern self- from non-self-antigens, pri-
marily through the establishment of central immune tolerance [
163
,
164
], thus acting as a
safeguard against autoimmunity. Moreover, molecular mimics, such as those utilized in
vaccine design, tend to confer a reduced risk of provoking autoimmune reactions. This
reduced risk can be attributed to the relatively lower immunological pressure imposed
by these mimics when compared to actual pathogens, which inherently exert additional
immune pressure due to the manifestation of the disease itself.
The regulatory T cells (Tregs) also play a pivotal role in modulating the immune
response on encountering the antigen (from the vaccine), ensuring its proportionality and
averting the development of autoimmunity [
161
]. These regulatory processes are further
reinforced by natural checkpoints orchestrated by cytokines and other signaling molecules.
These checkpoints serve as crucial regulators, fine-tuning the intensity and duration of
the immune response [
165
], thus mitigating the risk of overly aggressive reactions that
could harm the body tissues, instead of just the pathogen. Furthermore, cells can release
immunosuppressive signals, including TGF-beta and IL-10, effectively dampening immune
responses and preventing unwarranted reactivity against self-antigens [
166
,
167
]. Antigen-
Microorganisms 2023,11, 2300 18 of 25
presenting cells (APCs) are another player in the immune system that present foreign
antigens to immune cells, while self-antigens are less likely to incite a response [
168
].
As a result, the potential for cross-reactivity leading to autoimmune triggers is notably
diminished in the context of mimic-based vaccine design, thus underscoring the safety and
efficacy of this approach.
Additionally, the prudent approach of pre-vaccination testing for genetically predis-
posed individuals and the adoption of nanocarriers as alternatives to lipid adjuvants hold
promise in mitigating the risk of cross-reactivity and triggering autoimmunity [
163
,
169
].
Furthermore, comparative evaluations of diverse vaccine formulations, concerning their
capacity to induce or exacerbate pathology in relevant models, can yield valuable base-
line data about the efficacy and safety of these vaccines. The inclusion of comprehensive
immunological investigations, including autoimmune serology, within phases I to III of
clinical trials is warranted to holistically assess vaccine responses. Hence, mimic-based
innovative vaccine design, alongside the risk assessment and consideration of the inherent
protective mechanisms of the immune system, offers a promising pathway toward vaccines
that can effectively combat pathogens while sparing the human self-tissues from harm.
5. Conclusions
Investigation of autoimmune pathways associated with the identified human ho-
mologs of C. difficile revealed interesting connections to autoimmune diseases. The iden-
tified associations with autoimmune diseases, particularly rheumatoid arthritis, warrant
further investigation into the underlying mechanisms of autoimmunity and the specific
roles of these homologous proteins in disease pathogenesis. The structural similarity be-
tween human and C. difficile homologs suggests the possibility of using these bacterial
proteins as structural templates for vaccine design and development. Understanding the
conserved regions and functional motifs in these proteins may also aid in the design of
therapeutics targeting C. difficile and related human diseases. We fabricated a vaccine con-
struct using conserved, safe, and immunogenic mimics. It demonstrated good response in
silico, but computational predictions have limitations, and we imply experimental research
to complement or refute our findings.
Supplementary Materials:
The following supporting information can be downloaded at:
https://www.mdpi.com/article/10.3390/microorganisms11092300/s1, Figure S1: (A) 3D structure of
vaccine construct using Swiss-Model (B) Ramachandran plot of the model; Table S1: Non-allergenic
vaccine constructs against C. difficile; Table S2: 3D structure statistics of the vaccine construct, us-
ing various tools; Table S3: HLA and TLR receptor interaction statistics with the designed vaccine
construct. Non-bonded contacts can involve attractive forces, such as van der Waals interactions
and hydrophobic interactions, or repulsive forces, such as steric clashes. No disulphide bond was
detected in any interaction.
Author Contributions:
S.A., Z.B., M.M.M. and A.A. conceptualized the study. Z.B., M.A.A. and
H.H.A. performed the experiments. N.A.A., M.M.M. and S.A. curated the data. S.A., Z.B., M.M.M.
and A.A. wrote and edited the original draft. H.H.A., N.A.A. and M.A.A. reviewed and edited the
draft. A.A., N.A.A. and M.A.A. visualized the study. S.A., Z.B., M.M.M. and H.H.A. obtained the
funding. S.A., Z.B. and M.M.M. administered the project. M.M.M. supervised the study. All authors
have read and agreed to the published version of the manuscript.
Funding:
This research was funded by the Deanship of Scientific Research at Najran University
Research Support Program under the grant program code NU/DRP/MRC/12/5.
Data Availability Statement:
All the data used or generated in this study are provided as an accession
number or relevant information as tables in the manuscript.
Acknowledgments:
The authors are also thankful to the Deanship of Scientific Research at Na-
jran University for funding this work, under the General Research Funding program grant code
NU/DRP/MRC/12/5.
Conflicts of Interest: The authors declare that they have no conflict of interest.
Microorganisms 2023,11, 2300 19 of 25
References
1.
Martins, Y.C.; Jurberg, A.D.; Daniel-Ribeiro, C.T.J.M. Visiting Molecular Mimicry Once More: Pathogenicity, Virulence, and
Autoimmunity. Microorganisms 2023,11, 1472. [CrossRef] [PubMed]
2.
Hussein, H.M.; Rahal, E.A. The role of viral infections in the development of autoimmune diseases. Crit. Rev. Microbiol.
2019
,45,
394–412. [CrossRef] [PubMed]
3.
Topi, S.; Bottalico, L.; Charitos, I.A.; Colella, M.; Di Domenico, M.; Palmirotta, R.; Santacroce, L.J. Biomolecular Mechanisms of
Autoimmune Diseases and Their Relationship with the Resident Microbiota: Friend or Foe? Pathophysiology
2022
,29, 507–536.
[CrossRef]
4.
Guarneri, F. The Concept of Infection-Triggered Autoimmunity. In Role of Microorganisms in Pathogenesis and Management of
Autoimmune Diseases: Volume I: Liver, Skin, Thyroid, Rheumatic & Myopathic Diseases; Springer: Berlin/Heidelberg, Germany, 2022;
pp. 3–19.
5. Mobasheri, L.; Nasirpour, M.H.; Masoumi, E.; Azarnaminy, A.F.; Jafari, M.; Esmaeili, S.-A. SARS-CoV-2 triggering autoimmune
diseases. Cytokine 2022,154, 155873. [CrossRef]
6.
Fugger, L.; Jensen, L.T.; Rossjohn, J.J. Challenges, progress, and prospects of developing therapies to treat autoimmune diseases.
Cell 2020,181, 63–80. [CrossRef] [PubMed]
7.
Burbelo, P.D.; Iadarola, M.J.; Keller, J.M.; Warner, B.M. Autoantibodies targeting intracellular and extracellular proteins in
autoimmunity. Front. Immunol. 2021,12, 548469. [CrossRef] [PubMed]
8.
Cunningham, M.W. Molecular mimicry, autoimmunity, and infection: The cross-reactive antigens of group A streptococci and
their sequelae. Microbiol. Spectr. 2019,7, 20. [CrossRef]
9. Adiguzel, Y.J. Molecular mimicry between SARS-CoV-2 and human proteins. Autoimmun. Rev. 2021,20, 102791. [CrossRef]
10.
Venigalla, S.S.K.; Premakumar, S.; Janakiraman, V. A possible role for autoimmunity through molecular mimicry in alphavirus
mediated arthritis. Sci. Rep. 2020,10, 938. [CrossRef]
11. Joubert, B.; Dalmau, J. The role of infections in autoimmune encephalitides. Rev. Neurol. 2019,175, 420–426. [CrossRef]
12.
Cuan-Baltazar, Y.; Soto-Vega, E. Microorganisms associated to thyroid autoimmunity. Autoimmun. Rev.
2020
,19, 102614.
[CrossRef] [PubMed]
13.
Youssefi, M.; Tafaghodi, M.; Farsiani, H.; Ghazvini, K.; Keikha, M. Helicobacter pylori infection and autoimmune diseases. Is
there an association with systemic lupus erythematosus, rheumatoid arthritis, autoimmune atrophy gastritis and autoimmune
pancreatitis? A systematic review and meta-analysis study. J. Microbiol. Immunol. Infect.
2021
,54, 359–369. [CrossRef] [PubMed]
14.
Begum, S.; Aiman, S.; Ahmad, S.; Samad, A.; Almehmadi, M.; Allahyani, M.; Aljuaid, A.; Afridi, S.G.; Khan, A. Molecular
Mimicry Analyses Unveiled the Human Herpes Simplex and Poxvirus Epitopes as Possible Candidates to Incite Autoimmunity.
Pathogens 2022,11, 1362. [CrossRef]
15.
Rahman, N.; Begum, S.; Khan, A.; Afridi, S.G.; Sahibzada, M.U.K.; Atwah, B.; Alhindi, Z.; Khan, H. An insight in Salmonella
typhi associated autoimmunity candidates’ prediction by molecular mimicry. Comput. Biol. Med.
2022
,148, 105865. [CrossRef]
[PubMed]
16.
Mashraqi, M.M.; Alzamami, A.; Alturki, N.A.; Alshamrani, S.; Alshahrani, M.M.; Almasoudi, H.H.; Basharat, Z. Molecular
Mimicry Mapping in Streptococcus pneumoniae: Cues for Autoimmune Disorders and Implications for Immune Defense
Activation. Pathogens 2023,12, 857. [CrossRef]
17.
Gouttefangeas, C.; Klein, R.; Maia, A. The good and the bad of T cell cross-reactivity: Challenges and opportunities for novel
therapeutics in autoimmunity and cancer. Front. Immunol. 2023,14, 1212546. [CrossRef]
18.
Reynolds, S.; Rafeek, R.A.M.; Hamlin, A.; Lepletier, A.; Pandey, M.; Ketheesan, N.; Good, M. Streptococcus pyogenes vaccine
candidates do not induce autoimmune responses in a rheumatic heart disease model. NPJ Vaccines 2023,8, 9. [CrossRef]
19.
Burova, L.A.; Suvorov, A.N.; Pigarevsky, P.V.; Totolian, A.A. Autoimmune streptococcal glomerulonephritis: The problem of
nephritogenicity of Streptococcus pyogenes.Russ. J. Infect. Immun. 2023,13, 409–429. [CrossRef]
20.
Cutforth, T.; De Mille, M.M.; Agalliu, I.; Agalliu, D.J. CNS autoimmune disease after Streptococcus pyogenes infections: Animal
models, cellular mechanisms and genetic factors. Future Neurol. 2016,11, 63–76. [CrossRef]
21.
Draborg, A.H.; Duus, K.; Houen, G. Epstein-Barr virus in systemic autoimmune diseases. Clin. Dev. Immunol.
2013
,2013, 535738.
[CrossRef]
22.
Poole, B.D.; Scofield, R.H.; Harley, J.B.; James, J. Epstein-Barr virus and molecular mimicry in systemic lupus erythematosus.
Autoimmunity 2006,39, 63–70. [CrossRef] [PubMed]
23.
Jog, N.R.; James, J.A. Epstein Barr virus and autoimmune responses in systemic lupus erythematosus. Front. Immunol.
2021
,
11, 623944. [CrossRef] [PubMed]
24.
Chiba, T.; Goto, S.; Yokosuka, O.; Imazeki, F.; Tanaka, M.; Fukai, K.; Takahashi, Y.; Tsujimura, H.; Saisho, H. Fatal chronic active
Epstein–Barr virus infection mimicking autoimmune hepatitis. Eur. J. Gastroenterol. Hepatol. 2004,16, 225–228. [CrossRef]
25. Lünemann, J.D.; Münz, C. Epstein-Barr virus and multiple sclerosis. Nat. Rev. Microbiol. 2007,7, 253–258. [CrossRef]
26.
Paroli, M.; Iannucci, G.; Accapezzato, D. Hepatitis C virus infection and autoimmune diseases. Int. J. Gen. Med.
2012
,5, 903–907.
[CrossRef]
27.
Dudak, F.C.; Boyaci, I.H.; Orner, B.P. The discovery of small-molecule mimicking peptides through phage display. Molecules
2011
,
16, 774–789. [CrossRef]
Microorganisms 2023,11, 2300 20 of 25
28.
Trela, M.; Nelson, P.N.; Rylance, P.B. The role of molecular mimicry and other factors in the association of Human Endogenous
Retroviruses and autoimmunity. Apmis 2016,124, 88–104. [CrossRef] [PubMed]
29.
Zhou, L.; Feng, T.; Xu, S.; Gao, F.; Lam, T.T.; Wang, Q.; Wu, T.; Huang, H.; Zhan, L.; Li, L. ggmsa: A visual exploration tool for
multiple sequence alignment and associated data. Brief. Bioinform. 2022,23, bbac222. [CrossRef]
30.
Silva, L.; Antunes, A. Omics and Remote Homology Integration to Decipher Protein Functionality. In Homology Modeling: Methods
and Protocols; Springer: Berlin/Heidelberg, Germany, 2023; pp. 61–81.
31.
Lundegaard, C.; Lamberth, K.; Harndahl, M.; Buus, S.; Lund, O.; Nielsen, M. NetMHC-3.0: Accurate web accessible predictions of
human, mouse and monkey MHC class I affinities for peptides of length 8–11. Nucleic Acids Res.
2008
,36, W509–W512. [CrossRef]
32.
Vita, R.; Mahajan, S.; Overton, J.A.; Dhanda, S.K.; Martini, S.; Cantrell, J.R.; Wheeler, D.K.; Sette, A.; Peters, B. The immune
epitope database (IEDB): 2018 update. Nucleic Acids Res. 2019,47, D339–D343. [CrossRef]
33.
Morsy, S.; Morsy, A. Epitope mimicry analysis of SARS-CoV-2 surface proteins and human lung proteins. J. Mol. Graph. Model.
2021,105, 107836. [CrossRef] [PubMed]
34.
Lunemann, J.D.; Jelcic, I.; Roberts, S.; Lutterotti, A.; Tackenberg, B.; Martin, R.; Munz, C. EBNA1-specific T cells from patients
with multiple sclerosis cross react with myelin antigens and co-produce IFN-gamma and IL-2. J. Exp. Med.
2008
,205, 1763–1773.
[CrossRef]
35.
Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J. Characterizationof spike glycoprotein of SARS-CoV-2
on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020,11, 1620. [CrossRef] [PubMed]
36.
Kyrklund, M.; Bildo, M.; Akhi, R.; Nissinen, A.E.; Pussinen, P.; Hörkkö, S.; Wang, C. Humoral immune response to heat shock
protein 60 of Aggregatibacter actinomycetemcomitans and cross-reactivity with malondialdehyde acetaldehyde-modified LDL.
PLoS ONE 2020,15, e0230682. [CrossRef] [PubMed]
37.
Lawson, C.M. Evidence for mimicry by viral antigens in animal models of autoimmune disease including myocarditis. Cell. Mol.
Life Sci. 2000,57, 552–560. [CrossRef]
38.
Benvenga, S.; Guarneri, F. Molecular mimicry and autoimmune thyroid disease. Rev. Endocr. Metab. Disord.
2016
,17, 485–498.
[CrossRef]
39.
Amedei, A.; Bergman, M.P.; Appelmelk, B.J.; Azzurri, A.; Benagiano, M.; Tamburini, C.; van der Zee, R.; Telford, J.L.;
Vandenbroucke-Grauls, C.M.; D’Elios, M. Molecular mimicry between Helicobacter pylori antigens and H+, K+—adenosine
triphosphatase in human gastric autoimmunity. J. Exp. Med. 2003,198, 1147–1156. [CrossRef]
40.
Nora, T.; Lomma, M.; Gomez-Valero, L.; Buchrieser, C. Molecular mimicry: An important virulence strategy employed by
Legionella pneumophila to subvert host functions. Future Microbiol. 2009,4, 691–701. [CrossRef]
41.
Tanim, T.M. In Silico Identification of Molecular Mimicry Between T-Cell Epitopes of Borrelia Burgdorferi and the Human Proteome:
Implications for Autoimmune Response in Lyme Disease; Brac University: Dhaka, Bangladesh, 2022.
42.
Gkoutzourelas, A.; Barmakoudi, M.; Bogdanos, D. A bioinformatics analysis reveals novel pathogens as molecular mimicry
triggers of systemic sclerosis. Mediterr. J. Rheumatol. 2020,31, 50. [CrossRef]
43.
Pahari, S.; Chatterjee, D.; Negi, S.; Kaur, J.; Singh, B.; Agrewala, J. Morbid sequences suggest molecular mimicry between
microbial peptides and self-antigens: A possibility of inciting autoimmunity. Front. Microbiol. 2017,8, 1938. [CrossRef]
44.
Vardhini, D.; Suneetha, S.; Ahmed, N.; Joshi, D.; Karuna, S.; Magee, X.; Vijayalakshmi, D.; Sridhar, V.; Karunakar, K.;
Archelos, J.; et al. Comparative proteomics of the Mycobacterium leprae binding protein myelin P0: Its implication in leprosy and
other neurodegenerative diseases. Infect. Genet. Evol. 2004,4, 21–28. [CrossRef] [PubMed]
45.
Ludin, P.; Nilsson, D.; Mäser, P. Genome-wide identification of molecular mimicry candidates in parasites. PLoS ONE
2011
,
6, e17546. [CrossRef] [PubMed]
46.
Dreyfus, D.H.; Farina, A.; Farina, G. Molecular mimicry, genetic homology, and gene sharing proteomic “molecular fingerprints”
using an EBV (Epstein-Barr virus)-derived microarray as a potential diagnostic method in autoimmune disease. Immunol. Res.
2018,66, 686–695. [CrossRef] [PubMed]
47.
Repac, J.; Mandi´c, M.; Luni´c, T.; Boži´c, B.; Boži´c Nedeljkovi´c, B. Mining the capacity of human-associated microorganisms
to trigger rheumatoid arthritis—A systematic immunoinformatics analysis of T cell epitopes. PLoS ONE
2021
,16, e0253918.
[CrossRef]
48.
Garg, A.; Kumari, B.; Kumar, R.; Kumar, M. miPepBase: A database of experimentally verified peptides involved in molecular
mimicry. Front. Microbiol. 2017,8, 2053. [CrossRef]
49.
Martínez, Y.A.; Guo, X.; Portales-Pérez, D.P.; Rivera, G.; Castañeda-Delgado, J.E.; García-Pérez, C.A.; Enciso-Moreno, J.A.;
Lara-Ramírez, E. The analysis on the human protein domain targets and host-like interacting motifs for the MERS-CoV and
SARS-CoV/CoV-2 infers the molecular mimicry of coronavirus. PLoS ONE 2021,16, e0246901.
50.
Tayal, S.; Bhatia, V.; Mehrotra, T.; Bhatnagar, S. ImitateDB: A database for domain and motif mimicry incorporating host and
pathogen protein interactions. Amino Acids 2022,54, 923–934. [CrossRef]
51.
Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Tunyasuvunakool, K.; Ronneberger, O.; Bates, R.; Žídek, A.;
Bridgland, A.; et al
. AlphaFold 2. 2020. Available online: https://predictioncenter.org/casp14/doc/presentations/2020_1
2_01_TS_predictor_AlphaFold2.pdf (accessed on 14 June 2023).
52.
David, A.; Islam, S.; Tankhilevich, E.; Sternberg, M. The AlphaFold database of protein structures: A biologist’s guide. J. Mol. Biol.
2022,434, 167336. [CrossRef]
Microorganisms 2023,11, 2300 21 of 25
53.
Gelly, J.-C.; Joseph, A.P.; Srinivasan, N.; de Brevern, A. iPBA: A tool for protein structure comparison using sequence alignment
strategies. Nucleic Acids Res. 2011,39, W18–W23. [CrossRef]
54.
Zhang, Y.; Skolnick, J. TM-align: A protein structure alignment algorithm based on the TM-score. Nucleic Acids Res.
2005
,33,
2302–2309. [CrossRef]
55.
Rahmati, S.; Abovsky, M.; Pastrello, C.; Kotlyar, M.; Lu, R.; Cumbaa, C.A.; Rahman, P.; Chandran, V.; Jurisica, I. pathDIP 4: An
extended pathway annotations and enrichment analysis resource for human, model organisms and domesticated species. Nucleic
Acids Res. 2020,48, D479–D488. [CrossRef] [PubMed]
56.
Nguyen, D.-T.; Mathias, S.; Bologa, C.; Brunak, S.; Fernandez, N.; Gaulton, A.; Hersey, A.; Holmes, J.; Jensen, L.J.; Karlsson, A.
Pharos: Collating protein information to shed light on the druggable genome. Nucleic Acids Res.
2017
,45, D995–D1002. [CrossRef]
[PubMed]
57.
Rahmati, S.; Abovsky, M.; Pastrello, C.; Jurisica, I. pathDIP: An annotated resource for known and predicted human gene-pathway
associations and pathway enrichment analysis. Nucleic Acids Res. 2017,45, D419–D426. [CrossRef]
58.
Doytchinova, I.A.; Flower, D. VaxiJen: A server for prediction of protective antigens, tumour antigens and subunit vaccines. BMC
Bioinform. 2007,8, 1–7. [CrossRef] [PubMed]
59.
Gasteiger, E.; Hoogland, C.; Gattiker, A.; Duvaud, S.e.; Wilkins, M.R.; Appel, R.D.; Bairoch, A. Protein Identification and Analysis
Tools on the ExPASy Server; Springer: Berlin/Heidelberg, Germany, 2005.
60.
Grote, A.; Hiller, K.; Scheer, M.; Münch, R.; Nörtemann, B.; Hempel, D.C.; Jahn, D. JCat: A novel tool to adapt codon usage of a
target gene to its potential expression host. Nucleic Acids Res. 2005,33, W526–W531. [CrossRef] [PubMed]
61.
Castiglione, F.; Bernaschi, M. C-immsim: Playing with the immune response. In Proceedings of the Sixteenth International
Symposium on Mathematical Theory of Networks and Systems (MTNS2004), KU Leuven, Belgium, 5–9 July 2004.
62.
Jiang, S.; Bottazzi, M.E.; Du, L.; Lustigman, S.; Tseng, C.-T.K.; Curti, E.; Jones, K.; Zhan, B.; Hotez, P. Roadmap to developing a
recombinant coronavirus S protein receptor-binding domain vaccine for severe acute respiratory syndrome. Expert Rev. Vaccines
2012,11, 1405–1413. [CrossRef]
63.
Delgado, M.F.; Coviello, S.; Monsalvo, A.C.; Melendi, G.A.; Hernandez, J.Z.; Batalle, J.P.; Diaz, L.; Trento, A.; Chang, H.-Y.;
Mitzner, W. Lack of antibody affinity maturation due to poor Toll-like receptor stimulation leads to enhanced respiratory syncytial
virus disease. Nat. Med. 2009,15, 34–41. [CrossRef]
64.
Waterhouse, A.; Bertoni, M.; Bienert, S.; Studer, G.; Tauriello, G.; Gumienny, R.; Heer, F.T.; de Beer, T.A.P.; Rempfer, C.; Bordoli, L.
SWISS-MODEL: Homology modelling of protein structures and complexes. Nucleic Acids Res.
2018
,46, W296–W303. [CrossRef]
65. Zhang, Y. I-TASSER server for protein 3D structure prediction. BMC Bioinform. 2008,9, 1–8. [CrossRef]
66.
Kozakov, D.; Hall, D.R.; Xia, B.; Porter, K.A.; Padhorny, D.; Yueh, C.; Beglov, D.; Vajda, S. The ClusPro web server for protein–
protein docking. Nat. Protoc. 2017,12, 255–278. [CrossRef]
67.
Comeau, S.R.; Gatchell, D.W.; Vajda, S.; Camacho, C. ClusPro: A fully automated algorithm for protein–protein docking. Nucleic
Acids Res. 2004,32, W96–W99. [CrossRef] [PubMed]
68.
Xue, L.C.; Rodrigues, J.P.; Kastritis, P.L.; Bonvin, A.M.; Vangone, A.J.B. PRODIGY: A web server for predicting the binding affinity
of protein–protein complexes. Bioinformatics 2016,32, 3676–3678. [CrossRef]
69.
Loshaj-Shala, A.; Regazzoni, L.; Daci, A.; Orioli, M.; Brezovska, K.; Panovska, A.P.; Beretta, G.; Suturkova, L. Guillain Barré
syndrome (GBS): New insights in the molecular mimicry between C. jejuni and human peripheral nerve (HPN) proteins.
J. Neuroimmunol. 2015,289, 168–176. [CrossRef] [PubMed]
70.
Yokota, S.-i.; Chiba, S.; Furuyama, H.; Fujii, N. Cerebrospinal fluids containing anti-HSP70 autoantibodies from multiple sclerosis
patients augment HSP70-induced proinflammatory cytokine production in monocytic cells. J. Neuroimmunol.
2010
,218, 129–133.
[CrossRef]
71.
Mosenson, J.A.; Zloza, A.; Nieland, J.D.; Garrett-Mayer, E.; Eby, J.M.; Huelsmann, E.J.; Kumar, P.; Denman, C.J.; Lacek, A.T.;
Kohlhapp, F. Mutant HSP70 reverses autoimmune depigmentation in vitiligo. Sci. Transl. Med.
2013
,5, 174ra128. [CrossRef]
[PubMed]
72.
Lemos, J.; Giambiagi-Demarval, M.; Castro, A. Expression of heat-shock proteins in Streptococcus pyogenes and their immunoreac-
tivity with sera from patients with streptococcal diseases. J. Med. Microbiol. 1998,47, 711–715. [CrossRef] [PubMed]
73.
Shraibman, B.; Barnea, E.; Kadosh, D.M.; Haimovich, Y.; Slobodin, G.; Rosner, I.; López-Larrea, C.; Hilf, N.; Kuttruff, S.;
Song, C.; et al
. Identification of tumor antigens among the HLA peptidomes of glioblastoma tumors and plasma. Mol. Cell.
Proteom. 2018,17, 2132–2145. [CrossRef] [PubMed]
74.
Sudhir, P.-R.; Lin, T.-D.; Zhang, Q. HLA allele-specific quantitative profiling of type 1 diabetic b lymphocyte immunopeptidome.
J. Proteome Res. 2021,21, 250–264. [CrossRef] [PubMed]
75.
Shoda, H.; Hanata, N.; Sumitomo, S.; Okamura, T.; Fujio, K.; Yamamoto, K. Immune responses to Mycobacterial heat shock
protein 70 accompany self-reactivity to human BiP in rheumatoid arthritis. Sci. Rep. 2016,6, 22486. [CrossRef]
76.
Requena, J.M.; Soto, M.; Guzman, F.; Maekelt, A.; Noya, O.; Patarroyo, M.E.; Alonso, C. Mapping of antigenic determinants of the
T. cruzi hsp70 in chagasic and healthy individuals. Mol. Immunol. 1993,30, 1115–1121. [CrossRef]
77.
Adams, E.; Britton, W.; Morgan, A.; Sergeantson, S.; Basten, A. Individuals from Different Populations Identify Multiple and
Diverse T-Cell Determinants on Mycobacterial HSP70. Scand. J. Immunol. 1994,39, 588–596. [CrossRef] [PubMed]
78.
Sigdel, K.R.; Cheng, A.; Wang, Y.; Duan, L.; Zhang, Y. The emerging functions of long noncoding RNA in immune cells:
Autoimmune diseases. J. Immunol. Res. 2015,2015, 848790. [CrossRef] [PubMed]
Microorganisms 2023,11, 2300 22 of 25
79.
Read, S.A.; Tay, E.; Shahidi, M.; George, J.; Douglas, M. Hepatitis C virus infection mediates cholesteryl ester synthesis to facilitate
infectious particle production. J. Gen. Virol. 2014,95, 1900–1910. [CrossRef] [PubMed]
80.
Li, Y.; Leung, E.L.-H.; Pan, H.; Yao, X.; Huang, Q.; Wu, M.; Xu, T.; Wang, Y.; Cai, J.; Li, R. Identification of potential genetic causal
variants for rheumatoid arthritis by whole-exome sequencing. Oncotarget 2017,8, 111119. [CrossRef] [PubMed]
81.
Dudek, N.L.; Tan, C.T.; Gorasia, D.G.; Croft, N.P.; Illing, P.T.; Purcell, A. Constitutive and inflammatory immunopeptidome of
pancreatic β-cells. Diabetes 2012,61, 3018–3025. [CrossRef]
82.
Wong, S.M.; Jackson, M.D.; Akerley, B. Suppression of alternative lipooligosaccharide glycosyltransferase activity by UDP-
galactose epimerase enhances murine lung infection and evasion of serum IgM. Front. Cell. Infect. Microbiol.
2019
,9, 160.
[CrossRef]
83.
Maji, A.; Misra, R.; Kumar Mondal, A.; Kumar, D.; Bajaj, D.; Singhal, A.; Arora, G.; Bhaduri, A.; Sajid, A.; Bhatia, S. Expression
profiling of lymph nodes in tuberculosis patients reveal inflammatory milieu at site of infection. Sci. Rep.
2015
,5, 15214. [CrossRef]
84. Zhang, Z. Genomic biomarker heterogeneities between SARS-CoV-2 and COVID-19. Vaccines 2022,10, 1657. [CrossRef]
85.
Hu, J.; Zhou, S.; Guo, W. Construction of the coexpression network involved in the pathogenesis of thyroid eye disease via
bioinformatics analysis. Hum. Genom. 2022,16, 1–15. [CrossRef]
86.
Peng, S.; Wang, J.; Wei, S.; Li, C.; Zhou, K.; Hu, J.; Ye, X.; Yan, J.; Liu, W.; Gao, G. Endogenous cellular microRNAs mediate
antiviral defense against influenza A virus. Mol. Ther. Nucleic Acids 2018,10, 361–375. [CrossRef]
87.
Huang, T.; Huang, X.; Yao, M. miR-143 inhibits intracellular salmonella growth by targeting ATP6V1A in macrophage cells in pig.
Res. Vet. Sci. 2018,117, 138–143. [CrossRef] [PubMed]
88.
Liu, X.; Li, F.; Zhang, J.; Wang, L.; Wang, J.; Wen, Z.; Wang, Z.; Shuai, L.; Wang, X.; Ge, J. The ATPase ATP6V1A facilitates rabies
virus replication by promoting virion uncoating and interacting with the viral matrix protein. J. Biol. Chem.
2021
,296. [CrossRef]
[PubMed]
89.
Grodzki, M.; Bluhm, A.P.; Schaefer, M.; Tagmount, A.; Russo, M.; Sobh, A.; Rafiee, R.; Vulpe, C.D.; Karst, S.M.; Norris, M.
Genome-scale CRISPR screens identify host factors that promote human coronavirus infection. Genome Med.
2022
,14, 1–18.
[CrossRef]
90.
Blischak, J.D.; Tailleux, L.; Mitrano, A.; Barreiro, L.B.; Gilad, Y. Mycobacterial infection induces a specific human innate immune
response. Sci. Rep. 2015,5, 16882. [CrossRef] [PubMed]
91.
Raphael, I.; Webb, J.; Gomez-Rivera, F.; Chase Huizar, C.A.; Gupta, R.; Arulanandam, B.P.; Wang, Y.; Haskins, W.E.; Forsthuber, T.
Serum neuroinflammatory disease-induced central nervous system proteins predict clinical onset of experimental autoimmune
encephalomyelitis. Front. Immunol. 2017,8, 812. [CrossRef] [PubMed]
92.
Gerstner, C.; Dubnovitsky, A.; Sandin, C.; Kozhukh, G.; Uchtenhagen, H.; James, E.A.; Ronnelid, J.; Ytterberg, A.J.; Pieper, J.;
Reed, E. Functional and Structural Characterization of a Novel HLA-DRB1* 04: 01-Restricted alpha-Enolase T Cell Epitope in
Rheumatoid Arthritis. Front. Immunol. 2016,7, 494. [CrossRef] [PubMed]
93.
Moreno, I., Jr.; Rodríguez-Sánchez, I.; Schafer, X.; Munger, J. Human cytomegalovirus induces neuronal enolase to support virally
mediated metabolic remodeling. Proc. Natl. Acad. Sci. USA 2022,119, e2205789119. [CrossRef]
94.
Gaseitsiwe, S.; Valentini, D.; Mahdavifar, S.; Magalhaes, I.; Hoft, D.F.; Zerweck, J.; Schutkowski, M.; Andersson, J.; Reilly, M.;
Maeurer, M. Pattern recognition in pulmonary tuberculosis defined by high content peptide microarray chip analysis representing
61 proteins from M. tuberculosis.PLoS ONE 2008,3, e3840. [CrossRef]
95.
Zhao, M.; Wu, J.; Li, X.; Gao, Y. Urinary candidate biomarkers in an experimental autoimmune myocarditis rat model. J. Proteom.
2018,179, 71–79. [CrossRef]
96.
Mishra, N.; Huang, X.; Joshi, S.; Guo, C.; Ng, J.; Thakkar, R.; Wu, Y.; Dong, X.; Li, Q.; Pinapati, R. Immunoreactive peptide maps
of SARS-CoV-2. Commun. Biol. 2021,4, 225. [CrossRef]
97.
González-Sapienza, G.; Cachau, R. Identification of critical residues of an immunodominant region of Echinococcus granulosus
antigen B. J. Biol. Chem. 2003,278, 20179–20184. [CrossRef] [PubMed]
98. Zhang, P.; Minardi, L.M.; Todd Kuenstner, J.; Zekan, S.M.; Zhu, F.; Hu, Y.; Kruzelock, R. Cross—Reactivity of antibodies against
microbial proteins to human tissues as basis of Crohn’s disease and other autoimmune diseases. bioRxiv 2017, 116574.
99.
Zhang, P.; Minardi, L.M.; Kuenstner, J.T.; Zekan, S.M.; Kruzelock, R.J.B. Seroprevalence of anti-microbial antibodies in the normal
healthy population with implications in chronic diseases. bioRxiv 2019, 693655.
100.
Syed, S.; Viazmina, L.; Mager, R.; Meri, S.; Haapasalo, K. Streptococci and the complement system: Interplay during infection,
inflammation and autoimmunity. FEBS Lett. 2020,594, 2570–2585. [CrossRef] [PubMed]
101.
Harvey, K.L.; Jarocki, V.M.; Charles, I.G.; Djordjevic, S. The diverse functional roles of elongation factor Tu (EF-Tu) in microbial
pathogenesis. Front. Microbiol. 2019,10, 2351.
102.
Lo, K.C.; Sullivan, E.; Bannen, R.M.; Jin, H.; Rowe, M.; Li, H.; Pinapati, R.S.; Cartwright, A.J.; Tan, J.C.; Patel, J.; et al.
Comprehensive profiling of the rheumatoid arthritis antibody repertoire. Arthritis Rheumatol. 2020,72, 242–250. [CrossRef]
103.
Venema, W.J.; Hiddingh, S.; de Boer, J.H.; Claas, F.H.; Mulder, A.; den Hollander, A.I.; Stratikos, E.; Sarkizova, S.; van der
Veken, L.T.; Janssen, G. ERAP2 increases the abundance of a peptide submotif highly selective for the birdshot uveitis-associated
HLA-A29. Front. Immunol. 2021,12, 634441. [CrossRef]
104.
Caradonna, S.J.; Cheng, Y. Induction of uracil-DNA glycosylase and dUTP nucleotidohydrolase activity in herpes simplex
virus-infected human cells. J. Biol. Chem. 1981,256, 9834–9837. [CrossRef]
Microorganisms 2023,11, 2300 23 of 25
105.
Stuart, D.; Upton, C.; Higman, M.; Niles, E.; McFadden, G. A poxvirus-encoded uracil DNA glycosylase is essential for virus
viability. J. Virol. 1993,67, 2503–2512. [CrossRef]
106.
James, E.A.; Gates, T.J.; LaFond, R.E.; Yamamoto, S.; Ni, C.; Mai, D.; Gersuk, V.H.; O’Brien, K.; Nguyen, Q.-A.; Zeitner, B.
Neuroinvasive West Nile infection elicits elevated and atypically polarized T cell responses that promote a pathogenic outcome.
PLoS Pathog. 2016,12, e1005375. [CrossRef]
107.
Life, P.; Hassell, A.; Williams, K.; Young, S.; Bacon, P.; Southwood, T.; Gaston, J. Responses to gram negative enteric bacterial
antigens by synovial T cells from patients with juvenile chronic arthritis: Recognition of heat shock protein HSP60. J. Rheumatol.
1993,20, 1388–1396. [PubMed]
108.
Ford, P.; Gemmell, E.; Hamlet, S.; Hasan, A.; Walker, P.; West, M.; Cullinan, M.; Seymour, G. Cross-reactivity of GroEL antibodies
with human heat shock protein 60 and quantification of pathogens in atherosclerosis. Oral Microbiol. Immunol.
2005
,20, 296–302.
[CrossRef] [PubMed]
109.
Rodolico, V.; Tomasello, G.; Zerilli, M.; Martorana, A.; Pitruzzella, A.; Marino Gammazza, A.; David, S.; Zummo, G.; Damiani, P.;
Accomando, S.; et al. Hsp60 and Hsp10 increase in colon mucosa of Crohn’s disease and ulcerative colitis. Cell Stress Chaperones
2010,15, 877–884. [CrossRef] [PubMed]
110.
De Jong, H.; Lafeber, F.F.; de Jager, W.; Haverkamp, M.H.; Kuis, W.; Bijlsma, J.W.; Prakken, B.J.; Albani, S. PAN–DR-Binding
Hsp60 self epitopes induce an interleukin-10–mediated immune response in rheumatoid arthritis. Arthritis Rheum.
2009
,60,
1966–1976. [CrossRef] [PubMed]
111.
Dieudé, M.; Senécal, J.L.; Raymond, Y. Induction of endothelial cell apoptosis by heat-shock protein 60–reactive antibodies
from anti–endothelial cell autoantibody–positive systemic lupus erythematosus patients. Arthritis Rheum.
2004
,50, 3221–3231.
[CrossRef] [PubMed]
112.
Aragona, P.; Magazzu, G.; Macchia, G.; Bartolone, S.; Di Pasquale, G.; Vitali, C.; Ferreri, G. Presence of antibodies against
Helicobacter pylori and its heat-shock protein 60 in the serum of patients with Sjögren’s syndrome. J. Rheumatol.
1999
,26,
1306–1311.
113.
Jones, D.B.; Coulson, A.F.; Duff, G. Sequence homologies between hsp60 and autoantigens. Immunol. Today
1993
,14, 115–118.
[CrossRef]
114.
Marino Gammazza, A.; Rizzo, M.; Citarrella, R.; Rappa, F.; Campanella, C.; Bucchieri, F.; Patti, A.; Nikolic, D.; Cabibi, D.;
Amico, G.; et al. Elevated blood Hsp60, its structural similarities and cross-reactivity with thyroid molecules, and its presence on
the plasma membrane of oncocytes point to the chaperonin as an immunopathogenic factor in Hashimoto’s thyroiditis. Cell Stress
Chaperones 2014,19, 343–353. [CrossRef]
115.
Francesco, C.; Everly, C.; Alberto, J. HSPD1 (heat shock 60 kDa protein 1). Atlas Genet. Cytogenet. Oncol. Haematol.
2015
,19,
575–585.
116.
Vojdani, A.; Bazargan, M.; Vojdani, E.; Samadi, J.; Nourian, A.A.; Eghbalieh, N.; Cooper, E. Heat shock protein and gliadin peptide
promote development of peptidase antibodies in children with autism and patients with autoimmune disease. Clin. Vaccine
Immunol. 2004,11, 515–524. [CrossRef]
117.
Gonciarz, W.; Matusiak, A.; Rudnicka, K.; Rechci´nski, T.; Chałubi´nski, M.; Czkwianianc, E.; Broncel, M.; Gajewski, A.; Chmiela,
M. Autoantibodies to a specific peptide epitope of human Hsp60 (ATVLA) with homology to Helicobacter pylori HspB in H.
pylori-infected patients. APMIS J. Pathol. Microbiol. Immunol. 2019,127, 139–149.
118.
Lopez, J.A.M.; Cordero, J.F.B.; Russo, U.D.B.; Villarreal, M.A.G.; Hernández, F.A.T.; Bonilla, J.A.A.; Amaya, G.P.B.; Dumaguala,
M.R.J.; Valencia, C.C.R. Molecular Mimetism between Cystic Fibrosis and P. aeruginosa and S. aureus, Possible Cause of Autoim-
munity? In-Silico Analysis. J. Biomed. Sci. 2022,4, 2066–2072.
119.
Perschinka, H.; Mayr, M.; Millonig, G.; Mayerl, C.; van der Zee, R.; Morrison, S.G.; Morrison, R.P.; Xu, Q.; Wick, G. Cross-reactive
B-cell epitopes of microbial and human heat shock protein 60/65 in atherosclerosis. Arterioscler. Thromb. Vasc. Biol.
2003
,23,
1060–1065. [CrossRef] [PubMed]
120.
Abulafia-Lapid, R.; Elias, D.; Raz, I.; Keren-Zur, Y.; Atlan, H.; Cohen, I. T cell proliferative responses of type 1 diabetes patients
and healthy individuals to human hsp60 and its peptides. J. Autoimmun. 1999,12, 121–129. [CrossRef] [PubMed]
121.
Wizrah, M.S.; Chua, S.M.; Luo, Z.; Manik, M.K.; Pan, M.; Whyte, J.M.; Robertson, A.A.; Kappler, U.; Kobe, B.; Fraser, J.
AICAR transformylase/IMP cyclohydrolase (ATIC) is essential for de novo purine biosynthesis and infection by Cryptococcus
neoformans. J. Biol. Chem. 2022,298, 102453. [CrossRef] [PubMed]
122.
Guvenel, A.; Jozwik, A.; Ascough, S.; Ung, S.K.; Paterson, S.; Kalyan, M.; Gardener, Z.; Bergstrom, E.; Kar, S.; Habibi, M.
Epitope-specific airway-resident CD4+ T cell dynamics during experimental human RSV infection. J. Clin. Investig.
2020
,130,
523–538. [CrossRef]
123.
Cagnazzo, T.d.O.; Nogueira, C.T.; Castro, C.A.d.; Neris, D.M.; Fattori, A.C.M.; Correia, R.d.O.; Albuquerque, Y.R.; Fragelli,
B.D.d.L.; Mendes, T.M.F.; Allegretti, S. Investigating Immunization With Nucleotide Enzymes of Schistosoma Mansoni: Nucleo-
side Diphosphate Kinase and Adenylosuccinate Lyase as New Antigenic Targets Against Schistosomiasis. Front. Immunol.
2020
,
11, 569988. [CrossRef]
124.
Rahman, K.S.; Darville, T.; Russell, A.N.; O’Connell, C.M.; Wiesenfeld, H.C.; Hillier, S.L.; Chowdhury, E.U.; Juan, Y.-C.;
Kaltenboeck, B. Discovery of human-specific immunodominant Chlamydia trachomatis B cell epitopes. mSphere
2018
,3. [CrossRef]
125.
Lin, S.Y.-H.; Cheng, C.-W.; Su, E. Prediction of B-cell epitopes using evolutionary information and propensity scales. BMC
Bioinformatics 2013,14, S10. [CrossRef]
Microorganisms 2023,11, 2300 24 of 25
126.
Chauvin, A.; Bouvet, G.; Boulard, C. Humoral and cellular immune responses to Fasciola hepatica experimental primary and
secondary infection in sheep. Int. J. Parasitol. 1995,25, 1227–1241. [CrossRef]
127.
Boes, M. Role of natural and immune IgM antibodies in immune responses. Mol. Immunol.
2000
,37, 1141–1149. [CrossRef]
[PubMed]
128.
Coutelier, J.-P.; Van Der Logt, J.; Heessen, F. IgG subclass distribution of primary and secondary immune responses concomitant
with viral infection. J. Immunol. 1991,147, 1383–1386. [CrossRef] [PubMed]
129.
Guillerey, C.; Nakamura, K.; Vuckovic, S.; Hill, G.R.; Smyth, M. Immune responses in multiple myeloma: Role of the natural
immune surveillance and potential of immunotherapies. Cell. Mol. Life Sci. 2016,73, 1569–1589. [CrossRef] [PubMed]
130.
Round, J.L.; O’Connell, R.M.; Mazmanian, S. Coordination of tolerogenic immune responses by the commensal microbiota.
J. Autoimmun 2010,34, J220–J225. [CrossRef]
131.
Nikolich-Žugich, J. The twilight of immunity: Emerging concepts in aging of the immune system. Nat. Immunol.
2018
,19, 10–19.
[CrossRef]
132.
Siegrist, C. Vaccine Immunology. 2008, Volume 5, pp. 17–36. Available online: http://www.who.int/immunization/documents/
Elsevier_Vaccine_immunology.pdf (accessed on 16 June 2023).
133.
Souza-Fonseca-Guimaraes, F.; Adib-Conquy, M.; Cavaillon, J. Natural killer (NK) cells in antibacterial innate immunity: Angels
or devils? Mol. Med. 2012,18, 270–285. [CrossRef]
134.
Studer, G.; Rempfer, C.; Waterhouse, A.M.; Gumienny, R.; Haas, J.; Schwede, T. QMEANDisCo—Distance constraints applied on
model quality estimation. Bioinformatics 2020,36, 1765–1771. [CrossRef]
135.
Monirinasab, H.; Zakariazadeh, M.; Kohestani, H.; Kouhestani, M.; Fathi, F. Study of
β
-lactam-based drug interaction with
albumin protein using optical, sensing, and docking methods. J. Biol. Phys. 2022,48, 177–194. [CrossRef]
136.
Holler, P.D.; Kranz, D. Quantitative analysis of the contribution of TCR/pepMHC affinity and CD8 to T cell activation. Immunity
2003,18, 255–264. [CrossRef]
137.
Vangone, A.; Bonvin, A. Contacts-based prediction of binding affinity in protein–protein complexes. eLife
2015
,4, e07454.
[CrossRef]
138.
Czepiel, J.; Dró˙
zd˙
z, M.; Pituch, H.; Kuijper, E.J.; Perucki, W.; Mielimonka, A.; Goldman, S.; Wulta´nska, D.; Garlicki, A.; Biesiada,
G.; et al. Clostridium difficile infection. Eur. J. Clin. Microbiol. Infect. Dis. 2019,38, 1211–1221. [CrossRef]
139.
Balsells, E.; Shi, T.; Leese, C.; Lyell, I.; Burrows, J.; Wiuff, C.; Campbell, H.; Kyaw, M.H.; Nair, H. Global burden of Clostridium
difficile infections: A systematic review and meta-analysis. J. Glob. Health 2019,9, 010407. [CrossRef]
140.
Revolinski, S.L.; Munoz-Price, L. Clostridium difficile in immunocompromised hosts: A review of epidemiology, risk factors,
treatment, and prevention. Clin. Infect. Dis. 2019,68, 2144–2153. [CrossRef] [PubMed]
141.
Teneberg, S.; Lönnroth, I.; López, J.F.T.; Galili, U.; Halvarsson, M.Ö.; Ångström, J.; Karlsson, K. Molecular mimicry in the
recognition of glycosphingolipids by Gal
α
3Gal
β
4GlcNAc
β
-binding Clostridium difficile toxin A, human natural anti
α
-galactosyl
IgG and the monoclonal antibody Gal-13: Characterization of a binding-active human glycosphingolipid, non-identical with the
animal receptor. Glycobiology 1996,6, 599–609. [PubMed]
142.
Mindur, J.E.; Yadav, S.K.; Ito, N.; Senoh, M.; Kato, H.; Dhib-Jalbut, S.; Ito, K. Surface layer protein a expressed in Clostridioides
difficile DJNS06-36 possesses an encephalitogenic mimotope of myelin basic protein. Microorganisms
2020
,9, 34. [CrossRef]
[PubMed]
143.
Alam, J.; Kim, Y.C.; Choi, Y. Potential role of bacterial infection in autoimmune diseases: A new aspect of molecular mimicry.
Immune Netw. 2014,14, 7–13. [CrossRef]
144.
Mayo, I.; Arizti, P.; Parés, A.; Oliva, J.; Doforno, R.A.; de Sagarra, M.R.; Rodés, J.; Castaño, J. Antibodies against the COOH-
terminal region of E. coli ClpP protease in patients with primary biliary cirrhosis. J. Hepatol. 2000,33, 528–536.
145.
Zhang, C.; Zhang, T.-X.; Liu, Y.; Jia, D.; Zeng, P.; Du, C.; Yuan, M.; Liu, Q.; Wang, Y.; Shi, F. B-cell compartmental features and
molecular basis for therapy in autoimmune disease. Neurol. Neuroimmunol. Neuroinflamm. 2021,8, e1070. [CrossRef] [PubMed]
146.
Mansilla, M.J.; Montalban, X.; Espejo, C. Heat shock protein 70: Roles in multiple sclerosis. Mol. Med.
2012
,18, 1018–1028.
[CrossRef]
147.
Fouani, M.; Basset, C.A.; Mangano, G.D.; Leone, L.G.; Lawand, N.B.; Leone, A.; Barone, R. Heat shock proteins alterations in
rheumatoid arthritis. Int. J. Mol. Sci. 2022,23, 2806. [CrossRef]
148.
Lucchese, G.; Flöel, A. Chaperones. SARS-CoV-2 and Guillain-Barrésyndrome: Molecular mimicry with human heat shock
proteins as potential pathogenic mechanism. Cell Stress Chaperones 2020,25, 731–735. [CrossRef] [PubMed]
149.
Tukaj, S. Heat shock protein 70 as a double agent acting inside and outside the cell: Insights into autoimmunity. Int. J. Mol. Sci.
2020,21, 5298. [CrossRef] [PubMed]
150.
Yamamoto, T.; Eguchi, T. Heat Shock Proteins and Periodontitis–Cross-Reaction Between Bacterial and Human HSP in Periodontal
Infection Linking with Cardiovascular Diseases. In Heat Shock Proteins in Inflammatory Diseases; Springer: Berlin/Heidelberg,
Germany, 2020; pp. 19–32.
151.
Malik, J.A.; Lone, R. Heat shock proteins with an emphasis on HSP 60. Mol. Biol. Rep.
2021
,48, 6959–6969. [CrossRef] [PubMed]
152.
Repetto, O.; Zanussi, S.; Casarotto, M.; Canzonieri, V.; De Paoli, P.; Cannizzaro, R.; De Re, V. Differential proteomics of Helicobacter
pylori associated with autoimmune atrophic gastritis. Mol. Med. 2014,20, 57–71. [CrossRef] [PubMed]
153.
Qeshmi, S.A.; Dabbagh, F.; Haghighi, A.B.; Ghasemi, Y. Bioinformatics evaluation of the possibility of heat shock proteins as
autoantigens in multiple sclerosis based on molecular mimicry hypothesis. J. Neuroimmunol. 2016,295, 100–121. [CrossRef]
Microorganisms 2023,11, 2300 25 of 25
154.
Lee, Y.M.; Huh, K.C.; Yoon, S.M.; Jang, B.I.; Shin, J.E.; Koo, H.S.; Jung, Y.; Kim, S.H.; Moon, H.S.; Lee, S.; et al. Incidence and
clinical outcomes of Clostridium difficile infection after treatment with tuberculosis medication. Gut Liver
2016
,10, 250. [CrossRef]
155.
Kullin, B.R.; Reid, S.; Abratt, V. Clostridium difficile in patients attending tuberculosis hospitals in Cape Town, South Africa,
2014–2015. Afr. J. Lab. Med. 2018,7, 1–9. [CrossRef]
156.
Obuch-Woszczaty´nski, P.; Dubiel, G.; Harmanus, C.; Kuijper, E.; Duda, U.; Wulta´nska, D.; Van Belkum, A.; Pituch, H. Emergence
of Clostridium difficile infection in tuberculosis patients due to a highly rifampicin-resistant PCR ribotype 046 clone in Poland. Eur.
J. Clin. Microbiol. Infect. Dis. 2013,32, 1027–1030. [CrossRef]
157.
Chang, K.; Leung, C.; Yew, W.; Lam, F.; Ho, P.; Chau, C.; Cheng, V.; Yuen, K. Analyses of fluoroquinolones and Clostridium
difficile-associated diarrhoea in tuberculosis patients. Int. J. Tuberc. Lung Dis. 2009,13, 341–346.
158.
Collini, P.J.; Kuijper, E.; Dockrell, D. Clostridium difficile infection in patients with HIV/AIDS. Curr. HIV/AIDS Rep.
2013
,10,
273–282. [CrossRef]
159. Clem, A. Fundamentals of vaccine immunology. J. Glob. Infect. Dis. 2011,3, 73. [CrossRef] [PubMed]
160.
Irvine, D.J.; Swartz, M.A.; Szeto, G. Engineering synthetic vaccines using cues from natural immunity. Nat. Mater.
2013
,12,
978–990. [PubMed]
161.
Vadalà, M.; Poddighe, D.; Laurino, C.; Palmieri, B. Vaccination and autoimmune diseases: Is prevention of adverse health effects
on the horizon? EPMA J. 2017,8, 295–311. [PubMed]
162.
Horwitz, D.A.; Fahmy, T.M.; Piccirillo, C.A.; La Cava, A. Rebalancing immune homeostasis to treat autoimmune diseases. Trends
Immunol. 2019,40, 888–908. [CrossRef] [PubMed]
163.
Dangkoub, F.; Sankian, M.; Tafaghodi, M.; Jaafari, M.R.; Badiee, A. The impact of nanocarriers in the induction of antigen-specific
immunotolerance in autoimmune diseases. J. Control. Release 2021,339, 274–283. [CrossRef]
164.
Divangahi, M.; Aaby, P.; Khader, S.A.; Barreiro, L.B.; Bekkering, S.; Chavakis, T.; van Crevel, R.; Curtis, N.; DiNardo, A.R.;
Dominguez-Andres, J. Trained immunity, tolerance, priming and differentiation: Distinct immunological processes. Nat. Immunol.
2021,22, 2–6. [CrossRef]
165.
Chong, C.R.; Park, V.J.; Cohen, B.; Postow, M.A.; Wolchok, J.D.; Kamboj, M. Safety of inactivated influenza vaccine in cancer
patients receiving immune checkpoint inhibitors. Clin. Infect. Dis. 2020,70, 193–199. [CrossRef]
166.
Ke, Y.; Jiang, G.; Sun, D.; Kaplan, H.J.; Shao, H. Anti-CD3 antibody ameliorates experimental autoimmune uveitis by inducing
both IL-10 and TGF-βdependent regulatory T cells. Clin. Immunol. 2011,138, 311–320. [CrossRef]
167. Wraith, D. Therapeutic peptide vaccines for treatment of autoimmune diseases. Immunol. Lett. 2009,122, 134–136. [CrossRef]
168.
Ishina, I.A.; Zakharova, M.Y.; Kurbatskaia, I.N.; Mamedov, A.E.; Belogurov, A.A., Jr.; Gabibov, A. MHC Class II Presentation in
Autoimmunity. Cells 2023,12, 314.
169.
Liu, J.; Liu, Z.; Pang, Y.; Zhou, H. The interaction between nanoparticles and immune system: Application in the treatment of
inflammatory diseases. J. Nanobiotechnol. 2022,20, 1–25. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Acute post-streptococcal glomerulonephritis usually occurs as a complication after a streptococcal infection due to untimely or inadequate antibiotic therapy. The etiology of post-streptococcal glomerulonephritis has been studied rather comprehensively. Today, both clinicians and microbiologists do not deny the dominant role of Streptococcus pyogenes (streptococcus attributed to serological group A, GAS). Usually, emergence of acute post-streptococcal glomerulonephritis (APSGN) is associated with the so-called GAS-related "nephritogenicity" often judged by appearance and accumulation of antibodies to the antigens and extracellular products of streptococcal cells in patient blood. This interpretation is quite loose and most likely evidence about a link to the bacterial strain, rather than its nephritogenicity. Many studies refer and still attribute a leading role of "nephritogenic" factors to various streptococcal antigens and related biologically active products. Streptococcal nephritogenic factors include cross-reacting antigens, streptokinase, cysteine proteinase, endostreptosin a GAS cell membrane protein as well as plasmin-tropic enzyme glyceraldehyde-3-phosphate dehydrogenase. Nephritogenicity of all such streptococcal products is suspected to result from the fact that they are found in renal biopsies like specific patient blood serum antibodies. Regarding a term of nephritogenicity, it has been evidenced that it cannot be attributed to any specific streptococcal cell product. This review attempted to analyze a number of bacterial products as starting factors triggering this process. APSGN can be reproduced experimentally in rabbits by intravenous administration of a heat-killed Streptococcus pyogenes culture. In our experiments, strains of serotypes 1, 4, 12, 15, 22 were used. They produced M-proteins and had the ability to bind human and rabbit immunoglobulin G by interacting with the Fc part of the IgG molecule. In numerous series of experiments, evidence was obtained regarding the initiating role of GAS IgGFc-receptor proteins in developing APSGN. Recent studies confirmed the role of streptococcal IgGFc-binding proteins in the initiation of glomerulonephritis after animals were inoculated with temperature-killed IgGFc-positive GAS. This approach excluded a large group of bacterial extracellular agents from the list of APSGN-initiating candidates. An unconventional view on the pathogenesis of GAS-infection-coupled complications may allow approaching their prevention or new treatment strategies.
Article
Full-text available
T cells are main actors of the immune system with an essential role in protection against pathogens and cancer. The molecular key event involved in this absolutely central task is the interaction of membrane-bound specific T cell receptors with peptide-MHC complexes which initiates T cell priming, activation and recall, and thus controls a range of downstream functions. While textbooks teach us that the repertoire of mature T cells is highly diverse, it is clear that this diversity cannot possibly cover all potential foreign peptides that might be encountered during life. TCR cross-reactivity, i.e. the ability of a single TCR to recognise different peptides, offers the best solution to this biological challenge. Reports have shown that indeed, TCR cross-reactivity is surprisingly high. Hence, the T cell dilemma is the following: be as specific as possible to target foreign danger and spare self, while being able to react to a large spectrum of body-threatening situations. This has major consequences for both autoimmune diseases and cancer, and significant implications for the development of T cell-based therapies. In this review, we will present essential experimental evidence of T cell cross-reactivity, implications for two opposite immune conditions, i.e. autoimmunity vs cancer, and how this can be differently exploited for immunotherapy approaches. Finally, we will discuss the tools available for predicting cross-reactivity and how improvements in this field might boost translational approaches.
Article
Full-text available
Streptococcus pneumoniae contributes to a range of infections, including meningitis, pneumonia, otitis media, and sepsis. Infections by this bacterium have been associated with the phenomenon of molecular mimicry, which, in turn, may contribute to the induction of autoimmunity. In this study, we utilized a bioinformatics approach to investigate the potential for S. pneumoniae to incite autoimmunity via molecular mimicry. We identified 13 S. pneumoniae proteins that have significant sequence similarity to human proteins, with 11 of them linked to autoimmune disorders such as psoriasis, rheumatoid arthritis, and diabetes. Using in silico tools, we predicted the sequence as well as the structural homology among these proteins. Database mining was conducted to establish links between these proteins and autoimmune disorders. The antigenic, non-allergenic, and immunogenic sequence mimics were employed to design and validate an immune response via vaccine construct design. Mimic-based vaccine construct can prove effective for immunization against the S. pneumoniae infections. Immune response simulation and binding affinity was assessed through the docking of construct C8 to human leukocyte antigen (HLA) molecules and TLR4 receptor, with promising results. Additionally, these mimics were mapped as conserved regions on their respective proteins, suggesting their functional importance in S. pneumoniae pathogenesis. This study highlights the potential for S. pneumoniae to trigger autoimmunity via molecular mimicry and the possibility of vaccine design using these mimics for triggering defense response.
Article
Full-text available
The concept of molecular mimicry describes situations in which antigen sharing between parasites and hosts could benefit pathogen evasion from host immune responses. However, antigen sharing can generate host responses to parasite-derived self-like peptides, triggering autoimmunity. Since its conception, molecular mimicry and the consequent potential cross-reactivity following infections have been repeatedly described in humans, raising increasing interest among immunologists. Here, we reviewed this concept focusing on the challenge of maintaining host immune tolerance to self-components in parasitic diseases. We focused on the studies that used genomics and bioinformatics to estimate the extent of antigen sharing between proteomes of different organisms. In addition, we comparatively analyzed human and murine proteomes for peptide sharing with proteomes of pathogenic and non-pathogenic organisms. We conclude that, although the amount of antigenic sharing between hosts and both pathogenic and non-pathogenic parasites and bacteria is massive, the degree of this antigen sharing is not related to pathogenicity or virulence. In addition, because the development of autoimmunity in response to infections by microorganisms endowed with cross-reacting antigens is rare, we conclude that molecular mimicry by itself is not a sufficient factor to disrupt intact self-tolerance mechanisms.
Article
Full-text available
We have developed a candidate vaccine to protect against multiple strains of Streptococcus pyogenes infections. The candidate vaccine contains two synthetic peptides derived from S. pyogenes proteins: the M-protein epitope, p*17 and the IL-8 degrading S. pyogenes Cell-Envelope Proteinase (SpyCEP) epitope, K4S2. In this study we utilise a rat autoimmune valvulitis model that displays both the cardiac and neurobehavioural pathology associated with post-streptococcal sequelae, to assess if the vaccine candidate antigens induce autoimmune complications and inflammatory pathology. Each antigen was conjugated to carrier protein diphtheria toxoid (DT) and independently assessed for potential to induce autoimmune pathology in female Lewis rats. Rats were administered three subcutaneous doses, and one intranasal dose over a four-week study with a two-week recovery period. A positive control group received recombinant S. pyogenes M5 (rM5) protein, and the negative control group received PBS. Rats that received rM5 developed significant cardiac and neurological pathologies. There was no evidence of these pathologies in the PBS control group, or the rats administered either P*17-DT or K4S2-DT. This study provides further preclinical evidence of the safety of the vaccine candidates p*17 and K4S2 and their appropriateness as candidates in human clinical trials.
Article
Full-text available
Antigen presentation by major histocompatibility complex class II (MHC-II) molecules is crucial for eliciting an efficient immune response by CD4+ T cells and maintaining self-antigen tolerance. Some MHC-II alleles are known to be positively or negatively associated with the risk of the development of different autoimmune diseases (ADs), including those characterized by the emergence of autoreactive T cells. Apparently, the MHC-II presentation of self-antigens contributes to the autoimmune T cell response, initiated through a breakdown of central tolerance to self-antigens in the thymus. The appearance of autoreactive T cell might be the result of (i) the unusual interaction between T cell receptors (TCRs) and self-antigens presented on MHC-II; (ii) the posttranslational modifications (PTMs) of self-antigens; (iii) direct loading of the self-antigen to classical MHC-II without additional nonclassical MHC assistance; (iv) the proinflammatory environment effect on MHC-II expression and antigen presentation; and (v) molecular mimicry between foreign and self-antigens. The peculiarities of the processes involved in the MHC-II-mediated presentation may have crucial importance in the elucidation of the mechanisms of triggering and developing ADs as well as for clarification on the protective effect of MHC-II alleles that are negatively associated with ADs.
Article
Full-text available
Viruses depend on cellular metabolic resources to supply the energy and biomolecular building blocks necessary for their replication. Human cytomegalovirus (HCMV), a leading cause of birth defects and morbidity in immunosuppressed individuals, induces numerous metabolic activities that are important for productive infection. However, many of the mechanisms through which these metabolic activities are induced and how they contribute to infection are unclear. We find that HCMV infection of fibroblasts induces a neuronal gene signature as well as the expression of several metabolic enzyme isoforms that are typically expressed in other tissue types. Of these, the most substantially induced glycolytic gene was the neuron-specific isoform of enolase 2 (ENO2). Induction of ENO2 expression is important for HCMV-mediated glycolytic activation as well as for the virally induced remodeling of pyrimidine-sugar metabolism, which provides the glycosyl subunits necessary for protein glycosylation. Inhibition of ENO2 expression or activity reduced uridine diphosphate (UDP)-sugar pools, attenuated the accumulation of viral glycoproteins, and induced the accumulation of noninfectious viral particles. In addition, our data indicate that the induction of ENO2 expression depends on the HCMV UL38 protein. Collectively, our data indicate that HCMV infection induces a tissue atypical neuronal glycolytic enzyme to activate glycolysis and UDP-sugar metabolism, increase the accumulation of glycosyl building blocks, and enable the expression of an essential viral glycoprotein and the production of infectious virions.
Article
Full-text available
Clinical epidemiological studies have reported that viral infections cause autoimmune pathology in humans. Host-pathogen protein sequences and structure-based molecular mimicry cause autoreactive T cells to cross-activate. The aim of the current study was to implement im- munoinformatics approaches to infer sequence- and structure-based molecular mimicry between viral and human proteomic datasets. The protein sequences of all the so far known human-infecting viruses were obtained from the VIPR database, and complete human proteome data were retrieved from the NCBI repository. Based on a predefined, stringent threshold of comparative sequence analyses, 24 viral proteins were identified with significant sequence similarity to human proteins. PathDIP identified the enrichment of these homologous proteins in nine metabolic pathways with a p-value < 0.0001. Several viral and human mimic epitopes from these homologous proteins were predicted as strong binders of human HLA alleles, with IC50 < 50 nM. Downstream molecular docking analyses identified that lead virus-human homologous epitopes feasibly interact with HLA and TLR4 types of immune receptors. The vast majority of these top-hit homolog epitopic peptides belong to the herpes simplex and poxvirus families. These lead epitope biological sequences and 3D structural-based molecular mimicry may be promising for interpreting herpes simplex virus and poxvirus infection-mediated autoimmune disorders in humans.
Article
Full-text available
Genes functionally associated with SARS-CoV-2 infection and genes functionally related to the COVID-19 disease can be different, whose distinction will become the first essential step for successfully fighting against the COVID-19 pandemic. Unfortunately, this first step has not been completed in all biological and medical research. Using a newly developed max-competing logistic classifier, two genes, ATP6V1B2 and IFI27, stand out to be critical in the transcriptional response to SARS-CoV-2 infection with differential expressions derived from NP/OP swab PCR. This finding is evidenced by combining these two genes with another gene in predicting disease status to achieve better-indicating accuracy than existing classifiers with the same number of genes. In addition, combining these two genes with three other genes to form a five-gene classifier outperforms existing classifiers with ten or more genes. These two genes can be critical in fighting against the COVID-19 pandemic as a new focus and direction with their exceptional predicting accuracy. Comparing the functional effects of these genes with a five-gene classifier with 100% accuracy identified and tested from blood samples in our earlier work, the genes and their transcriptional response and functional effects on SARS-CoV-2 infection, and the genes and their functional signature patterns on COVID-19 antibodies, are significantly different. We will use a total of fourteen cohort studies (including breakthrough infections and omicron variants) with 1481 samples to justify our results. Such significant findings can help explore the causal and pathological links between SARS-CoV-2 infection and the COVID-19 disease, and fight against the disease with more targeted genes, vaccines, antiviral drugs, and therapies.
Chapter
In the recent years, several “omics” technologies based on specific biomolecules (from DNA, RNA, proteins, or metabolites) have won growing importance in the scientific field. Despite each omics possess their own laboratorial protocols, they share a background of bioinformatic tools for data integration and analysis. A recent subset of bioinformatic tools, based on available templates or remote homology protocols, allow computational fast and high-accuracy prediction of protein structures. The quickly predict of actually unsolved protein structures, together with late omics findings allow a boost of scientific advances in multiple fields such as cancer, longevity, immunity, mitochondrial function, toxicology, drug design, biosensors, and recombinant protein engineering. In this chapter, we assessed methodological approaches for the integration of omics and remote homology inferences to decipher protein functionality, opening the door to the next era of biological knowledge.Key wordsBioinformaticsComparative genomicsNatural selectionProtein functionRemote homologySpecies evolution