Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67.103
Filter
1.
PLoS One ; 19(9): e0308396, 2024.
Article in English | MEDLINE | ID: mdl-39283838

ABSTRACT

The American manatee (Trichechus manatus), experiencing population declines due to various threats, is the focus of conservation efforts that include the capture, rehabilitation, and release of orphaned calves when their mothers are unable to care for them. These efforts are compromised by the use of commercially available milk substitutes that lack essential components found in natural manatee breast milk, particularly immunoglobulin A (IgA). IgA plays a crucial role in nurturing the immune mucosal system and fostering a healthy microbiota. However, research on IgA in non-maternally fed manatees is limited due to the lack of species-specific reagents. To address this gap, our study employs immuno-informatics analysis to compare IgA sequences from manatees with those from other species, aiming to explore epitope similarity and sharing. We compared the protein sequence of manatee IgA with available IgA sequences, assessing similarity at the sequence, 3D structures, and epitope levels. Our findings reveal that human IgA exhibits the highest similarity in terms of sequence and 3D structure. Additionally, epitope analysis shows high conservation, identity, and similarity of predicted epitopes compared to human IgA. Future studies should focus on functional analysis using human IgA polyclonal reagents to detect manatee IgA in breast milk. Our findings highlight the potential of comparative analysis in advancing the understanding of immunology in non-human animals and overcoming challenges associated with the scarcity of species-specific reagents.


Subject(s)
Epitopes , Immunoglobulin A , Animals , Humans , Immunoglobulin A/immunology , Epitopes/immunology , Computational Biology/methods , Amino Acid Sequence
2.
PLoS Pathog ; 20(9): e1012042, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39250525

ABSTRACT

The generation of broadly neutralizing antibodies (bnAbs) to conserved epitopes on HIV Envelope (Env) is one of the cornerstones of HIV vaccine research. The animal models commonly used for HIV do not reliably produce a potent broadly neutralizing serum antibody response, with the exception of cows. Cows have previously produced a CD4 binding site response by homologous prime and boosting with a native-like Env trimer. In small animal models, other engineered immunogens were shown to focus antibody responses to the bnAb V2-apex region of Env. Here, we immunized two groups of cows (n = 4) with two regimens of V2-apex focusing Env immunogens to investigate whether antibody responses could be generated to the V2-apex on Env. Group 1 was immunized with chimpanzee simian immunodeficiency virus (SIV)-Env trimer that shares its V2-apex with HIV, followed by immunization with C108, a V2-apex focusing immunogen, and finally boosted with a cross-clade native-like trimer cocktail. Group 2 was immunized with HIV C108 Env trimer followed by the same HIV trimer cocktail as Group 1. Longitudinal serum analysis showed that one cow in each group developed serum neutralizing antibody responses to the V2-apex. Eight and 11 bnAbs were isolated from Group 1 and Group 2 cows, respectively, and showed moderate breadth and potency. Potent and broad responses in this study developed much later than previous cow immunizations that elicited CD4bs bnAbs responses and required several different immunogens. All isolated bnAbs were derived from the ultralong CDRH3 repertoire. The finding that cow antibodies can target more than one broadly neutralizing epitope on the HIV surface reveals the generality of elongated structures for the recognition of highly glycosylated proteins. The exclusive isolation of ultralong CDRH3 bnAbs, despite only comprising a small percent of the cow repertoire, suggests these antibodies outcompete the long and short CDRH3 antibodies during the bnAb response.


Subject(s)
AIDS Vaccines , Antibodies, Neutralizing , HIV Antibodies , HIV-1 , env Gene Products, Human Immunodeficiency Virus , Animals , Cattle , HIV Antibodies/immunology , AIDS Vaccines/immunology , HIV-1/immunology , Antibodies, Neutralizing/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Epitopes/immunology , HIV Infections/immunology , HIV Infections/prevention & control , Female , Immunization , Humans , Broadly Neutralizing Antibodies/immunology , Simian Immunodeficiency Virus/immunology
3.
PLoS One ; 19(9): e0310703, 2024.
Article in English | MEDLINE | ID: mdl-39325755

ABSTRACT

The resurgence of the Nipah virus (NiV) in 2023 has raised concerns for another potentially severe pandemic, given its history of high mortality from previous outbreaks. Unfortunately, no therapeutics and vaccines have been available for the virus. This study used immunoinformatics and molecular modeling to design and evaluate a multi-epitope subunit vaccine targeting NiV. The designed vaccine construct aims to stimulate immune responses in humans and two other intermediate animal hosts of the virus-swine and equine. Using several epitope prediction tools, ten peptides that induced B-lymphocyte responses, 17 peptides that induced cytotoxic T-lymphocyte (CTL) responses, and 12 peptides that induced helper T-lymphocyte (HTL) responses were mapped from nine NiV protein sequences. However, the CTL and HTL-inducing peptides were reduced to ten and eight, respectively, following molecular docking and dynamics. These screened peptides exhibited stability with 30 common major histocompatibility complex (MHC) receptors found in humans, swine, and equine. All peptides were linked using peptide linkers to form the multi-epitope construct and various adjuvants were tested to enhance its immunogenicity. The vaccine construct with resuscitation-promoting factor E (RpfE) adjuvant was selected as the final design based on its favorable physicochemical properties and superior immune response profile. Molecular docking was used to visualize the interaction of the vaccine to toll-like receptor 4 (TLR4), while molecular dynamics confirmed the structural stability of this interaction. Physicochemical property evaluation and computational simulations showed that the designed vaccine construct exhibited favorable properties and elicited higher antibody titers than the six multi-epitope NiV vaccine designs available in the literature. Further in vivo and in vitro experiments are necessary to validate the immunogenicity conferred by the designed vaccine construct and its epitope components. This study demonstrates the capability of computational methodologies in rational vaccine design and highlights the potential of cross-species vaccination strategies for mitigating potential NiV threats.


Subject(s)
Computational Biology , Henipavirus Infections , Nipah Virus , Vaccines, Subunit , Viral Vaccines , Nipah Virus/immunology , Animals , Vaccines, Subunit/immunology , Humans , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Viral Vaccines/immunology , Horses/immunology , Molecular Docking Simulation , Swine , Computer Simulation , Vaccination , Epitopes, T-Lymphocyte/immunology , Models, Molecular , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/immunology , Mice , Epitopes, B-Lymphocyte/immunology , Epitopes/immunology , Epitopes/chemistry , Immunoinformatics
4.
Nat Commun ; 15(1): 8318, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333495

ABSTRACT

Autoimmune attack toward pancreatic ß cells causes permanent loss of glucose homeostasis in type 1 diabetes (T1D). Insulin secretory granules store and secrete insulin but are also thought to be tissue messengers for T1D. Here, we show that the crinophagic granules (crinosome), a minor set of vesicles formed by fusing lysosomes with the conventional insulin dense-core granules (DCG), are pathogenic in T1D development in mouse models. Pharmacological inhibition of crinosome formation in ß cells delays T1D progression without affecting the dominant DCGs. Mechanistically, crinophagy inhibition diminishes the epitope repertoire in pancreatic islets, including cryptic, modified and disease-relevant epitopes derived from insulin. These unconventional insulin epitopes are largely undetectable in the MHC-II epitope repertoire of the thymus, where only canonical insulin epitopes are presented. CD4+ T cells targeting unconventional insulin epitopes display autoreactive phenotypes, unlike tolerized T cells recognizing epitopes presented in the thymus. Thus, the crinophagic pathway emerges as a tissue-intrinsic mechanism that transforms insulin from a signature thymic self-protein to a critical autoantigen by creating a peripheral-thymic mismatch in the epitope repertoire.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Insulin , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Mice , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Insulin/immunology , Epitopes/immunology , CD4-Positive T-Lymphocytes/immunology , Secretory Vesicles/metabolism , Secretory Vesicles/immunology , Mice, Inbred NOD , Autoantigens/immunology , Autoantigens/metabolism , Female , Disease Models, Animal , Thymus Gland/immunology , Humans , Lysosomes/metabolism , Lysosomes/immunology
5.
Nat Commun ; 15(1): 8382, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333507

ABSTRACT

We describe a process for rapid antibody affinity optimization by repertoire mining to identify clones across B cell clonal lineages based on convergent immune responses where antigen-specific clones with the same heavy (VH) and light chain germline segment pairs, or parallel lineages, bind a single epitope on the antigen. We use this convergence framework to mine unique and distinct VH lineages from rat anti-triggering receptor on myeloid cells 2 (TREM2) antibody repertoire datasets with high diversity in the third complementarity-determining loop region (CDR H3) to further affinity-optimize a high-affinity agonistic anti-TREM2 antibody while retaining critical functional properties. Structural analyses confirm a nearly identical binding mode of anti-TREM2 variants with subtle but significant structural differences in the binding interface. Parallel lineage repertoire mining is uniquely tailored to rationally explore the large CDR H3 sequence space in antibody repertoires and can be easily and generally applied to antibodies discovered in vivo.


Subject(s)
Antibody Affinity , Complementarity Determining Regions , Receptors, Immunologic , Animals , Complementarity Determining Regions/immunology , Antibody Affinity/immunology , Humans , Rats , Receptors, Immunologic/immunology , Receptors, Immunologic/genetics , Membrane Glycoproteins/immunology , B-Lymphocytes/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/genetics , Epitopes/immunology , Antibodies, Monoclonal/immunology , Antibodies/immunology
6.
PLoS Pathog ; 20(9): e1012550, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39321193

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel tick-borne bunyavirus that causes severe fever with thrombocytopenia syndrome (SFTS), with a high mortality rate of up to 30%. The envelope glycoproteins of SFTSV, glycoprotein N (Gn) and glycoprotein C (Gc), facilitate the recognition of host receptors and the process of membrane fusion, allowing the virus to enter host cells. We previously reported a monoclonal antibody, mAb 40C10, capable of neutralizing different genotypes of SFTSV and SFTSV-related viruses. However, the specific neutralization mechanism is poorly understood. In this study, we elucidated the high-resolution structure of the SFTSV Gn head domain in complex with mAb 40C10, confirming that the binding epitope in the domain I region of SFTSV Gn, and it represented that a novel binding epitope of SFTSV Gn was identified. Through in-depth structural and sequence analyses, we found that the binding sites of mAb 40C10 are relatively conserved among different genotypes of SFTSV and SFTSV-related Heartland virus and Guertu virus, elucidating the molecular mechanism underlying the broad-spectrum neutralizing activity of mAb 40C10. Furthermore, we humanized of mAb 40C10, which is originally of murine origin, to reduce its immunogenicity. The resulting nine humanized antibodies maintained potent affinity and neutralizing activity. One of the humanized antibodies exhibited neutralizing activity at picomolar IC50 values and demonstrated effective therapeutic and protective effects in a mouse infection model. These findings provide a novel target for the future development of SFTSV vaccines or drugs and establish a foundation for the research and development of antibody therapeutics for clinical applications.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Phlebovirus , Humans , Animals , Antibodies, Viral/immunology , Phlebovirus/immunology , Mice , Antibodies, Neutralizing/immunology , Severe Fever with Thrombocytopenia Syndrome/immunology , Viral Envelope Proteins/immunology , Antibodies, Monoclonal/immunology , Epitopes/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Broadly Neutralizing Antibodies/immunology
7.
Cell Rep ; 43(9): 114708, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39243373

ABSTRACT

Lassa fever continues to be a major public health burden in West Africa, yet effective therapies or vaccines are lacking. The isolation of protective neutralizing antibodies against the Lassa virus glycoprotein complex (GPC) justifies the development of vaccines that can elicit strong neutralizing antibody responses. However, Lassa vaccine candidates have generally been unsuccessful at doing so, and the associated antibody responses to these vaccines remain poorly characterized. Here, we establish an electron microscopy-based epitope mapping workflow that enables high-resolution structural characterization of polyclonal antibodies to the GPC. By applying this method to rabbits vaccinated with a recombinant GPC vaccine and a GPC-derived virus-like particle, we reveal determinants of neutralization that involve epitopes of the GPC-A competition cluster. Furthermore, by identifying undescribed immunogenic off-target epitopes, we expose the challenges that recombinant GPC vaccines face. By enabling detailed polyclonal antibody characterization, our work ushers in a next generation of more rational Lassa vaccine design.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Lassa Fever , Lassa virus , Lassa virus/immunology , Animals , Antibodies, Neutralizing/immunology , Rabbits , Antibodies, Viral/immunology , Lassa Fever/immunology , Lassa Fever/prevention & control , Epitopes/immunology , Viral Vaccines/immunology , Humans , Epitope Mapping , Antibody Formation/immunology
8.
Emerg Microbes Infect ; 13(1): 2396864, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39331815

ABSTRACT

Single B cells-based antibody platforms offer an effective approach for the discovery of useful antibodies for therapeutic or research purposes. Here we present a method for screening equine immunoglobins F(ab)2, which offers the potential advantage of reacting with multiple epitopes on the virus. Using equine influenza virus (EIV) as model, a hemagglutinin (HA) trimer was constructed to bait B cells in vaccinated horses. We screened 370 HA-specific B cells from 1 × 106 PBMCs and identified a diverse set of equine variable region gene sequences of heavy and light chains and then recombined with humanized Ig Fc. Recombinant equine Ig was then self-assembled in co-transfected 293 T cells, and subsequently optimized to obtain HA binding B-cell receptor (s). The recombinant antibodies exhibited a high binding affinity to the HA protein. Antibody H81 exhibited the highest cross neutralizing activities against EIV strains in vitro. Furthermore, it effectively protected EIV-challenged mice, resulting in significantly improved survival, reduced pulmonary inflammation and decreased viral titers. In silico predication identified a functional region of H81 comprising 27 key amino acids cross the main circulating EIV strains. The 12 amino acid residues in this region with the highest binding affinities were screened. Notably, the predicted epitopes of H81 encompassed the documented equine HA receptor binding site, validating its cross-neutralization. In summary, a rapid platform was successfully established to investigate the profiling of equine antigen-recognizing receptors (BCRs) following infection. This platform has the potential to optimize the screening of virus-neutralizing antibodies and aid in vaccine design.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , B-Lymphocytes , Orthomyxoviridae Infections , Animals , Horses , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Mice , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Antibodies, Neutralizing/immunology , Humans , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H3N8 Subtype/immunology , Influenza A Virus, H3N8 Subtype/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/genetics , Cross Reactions , HEK293 Cells , Influenza Vaccines/immunology , Immunologic Memory , Female , Epitopes/immunology
9.
Sci Rep ; 14(1): 20813, 2024 09 06.
Article in English | MEDLINE | ID: mdl-39242814

ABSTRACT

During pregnancy the immune system needs to maintain immune tolerance of the foetus while also responding to infection, which can cause premature activation of the inflammatory pathways leading to the onset of labour and preterm birth. The vaginal microbiome is an important modifier of preterm birth risk, with Lactobacillus dominance during pregnancy associated with term delivery while high microbial diversity is associated with an increased risk of preterm birth. Glycans on glycoproteins along the lower female reproductive tract are fundamental to microbiota-host interactions and the mediation of inflammatory responses. However, the specific glycan epitopes involved in these processes are not well understood. To address this, we conducted glycomic analyses of cervicovaginal fluid (CVF) from 36 pregnant women at high risk of preterm birth and 4 non-pregnant women. Our analysis of N- and O-glycans revealed a rich CVF glycome. While O-glycans were shown to be the main carriers of ABO blood group epitopes, the main features of N-glycans were the presence of abundant paucimannose and high mannose glycans, and a remarkable diversity of complex bi-, tri-, and tetra-antennary glycans decorated with fucose and sialic acid. We identified immuno-regulatory epitopes, such as Lewis antigens, and found that fucosylation was negatively correlated to pro-inflammatory factors, such as IL-1ß, MMP-8, C3a and C5a, while glycans with only sialylated antennae were mainly positively correlated to those. Similarly, paucimannose glycans showed a positive correlation to pro-inflammatory factors. We revealed a high abundance of glycans which have previously been identified as hallmarks of cancer and viral glycosylation, such as Man8 and Man9 high mannose glycans. Although each pregnant woman had a unique glycomic profile, longitudinal studies showed that the main glycosylation features were consistent throughout pregnancy in women who delivered at term, whereas women who experienced extreme preterm birth exhibited sharp changes in the CVF glycome shortly before delivery. These findings shed light on the processes underlying the role of glycosylation in maintaining a healthy vaginal microbiome and associated host immune responses. In addition, these discoveries facilitate our understanding of the lower female reproductive tract which has broad implications for women's health.


Subject(s)
Epitopes , Glycomics , Polysaccharides , Premature Birth , Vagina , Humans , Female , Premature Birth/immunology , Premature Birth/metabolism , Pregnancy , Glycosylation , Vagina/immunology , Vagina/metabolism , Vagina/microbiology , Adult , Epitopes/immunology , Polysaccharides/metabolism , Polysaccharides/immunology , Cervix Uteri/immunology , Cervix Uteri/metabolism , Body Fluids/immunology , Body Fluids/metabolism , Microbiota/immunology
10.
Front Immunol ; 15: 1432968, 2024.
Article in English | MEDLINE | ID: mdl-39247202

ABSTRACT

Helicobacter pylori (H. pylori) is an infectious bacterium that colonizes the stomach of approximately half of the global population. It has been classified as a Group I carcinogen by the World Health Organization due to its strong association with an increased incidence of gastric cancer and exacerbation of stomach diseases. The primary treatment for H. pylori infection currently involves triple or quadruple therapy, primarily consisting of antibiotics and proton pump inhibitors. However, the increasing prevalence of antibiotic resistance poses significant challenges to this approach, underscoring the urgent need for an effective vaccine. In this study, a novel multi-epitope H. pylori vaccine was designed using immunoinformatics. The vaccine contains epitopes derived from nine essential proteins. Software tools and online servers were utilized to predict, evaluate, and analyze the physiochemical properties, secondary and tertiary structures, and immunogenicity of the candidate vaccine. These comprehensive assessments ultimately led to the formulation of an optimal design scheme for the vaccine. Through constructing a novel multi-epitope vaccine based on immunoinformatics, this study offers promising prospects and great potential for the prevention of H. pylori infection. This study also provides a reference strategy to develop multi-epitope vaccines for other pathogens.


Subject(s)
Bacterial Vaccines , Computational Biology , Helicobacter Infections , Helicobacter pylori , Helicobacter pylori/immunology , Bacterial Vaccines/immunology , Computational Biology/methods , Helicobacter Infections/immunology , Helicobacter Infections/prevention & control , Humans , Antigens, Bacterial/immunology , Epitopes/immunology , Vaccine Development , Immunoinformatics
11.
Biochemistry (Mosc) ; 89(7): 1260-1272, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39218023

ABSTRACT

COVID-19 has caused millions of deaths and many times more infections worldwide, emphasizing the unpreparedness of the global health system in the face of new infections and the key role for vaccines and therapeutics, including virus-neutralizing antibodies, in prevention and containment of the disease. Continuous evolution of the SARS-CoV-2 coronavirus has been causing its new variants to evade the action of the immune system, which highlighted the importance of detailed knowledge of the epitopes of already selected potent virus-neutralizing antibodies. A single-chain antibody ("nanobody") targeting the SARS-CoV-2 receptor-binding domain (RBD), clone P2C5, had exhibited robust virus-neutralizing activity against all SARS-CoV-2 variants and, being a major component of the anti-COVID-19 formulation "GamCoviMab", had successfully passed Phase I of clinical trials. However, after the emergence of the Delta and XBB variants, a decrease in the neutralizing activity of this nanobody was observed. Here we report on the successful crystal structure determination of the RBD:P2C5 complex at 3.1 Å, which revealed the intricate protein-protein interface, sterically occluding full ACE2 receptor binding by the P2C5-neutralized RBD. Moreover, the structure revealed the developed RBD:P2C5 interface centered around residues Leu452 and Phe490, thereby explaining the evasion of the Delta or Omicron XBB, but not Omicron B.1.1.529 variant, as a result of the single L452R or F490S mutations, respectively, from the action of P2C5. The structure obtained is expected to foster nanobody engineering in order to rescue neutralization activity and will facilitate epitope mapping for other neutralizing nanobodies by competition assays.


Subject(s)
Antibodies, Neutralizing , SARS-CoV-2 , Single-Domain Antibodies , Spike Glycoprotein, Coronavirus , SARS-CoV-2/immunology , SARS-CoV-2/drug effects , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/chemistry , Humans , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/virology , Protein Domains , Protein Binding , Epitopes/immunology , Epitopes/chemistry , Models, Molecular , Immune Evasion , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/immunology , Binding Sites
12.
Parasitol Res ; 123(9): 314, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225716

ABSTRACT

Trypanosoma evansi is a unicellular protozoan responsible for causing a disease known as "surra," which is found in different regions of the world and primarily affects horses and camels. Few information is known about virulence factors released from the parasite within the animals. The organism can secrete extracellular vesicles (EVs), which transport a variety of molecules, including proteins. Before being considered exclusively as a means for eliminating unwanted substances, extracellular vesicles (EVs) have emerged as key players in intercellular communication, facilitating interactions between cells, host cells, and parasites, and even between parasites themselves. Thus, they may be used as potential biomarkers. This study aimed to assess the induction of EVs production by Ca+2, conduct a proteomic analysis of the EVs released by T. evansi, and identify epitopes that could serve as biomarkers. The findings indicated that Ca+2 is not an effective promoter of vesiculation in T. evansi. Furthermore, the proteomic analysis has identified multiple proteins that have been investigated as biomarkers or vaccine antigens, previously. A total of 442 proteins were identified, with 7 of them specifically recognizing 9 epitopes that are unique to T. evansi. At least one of these epitopes of TevSTIB805.9.11580 have been previously identified, which increases the possibility of further investigating its potential as a biomarker.


Subject(s)
Extracellular Vesicles , Proteomics , Protozoan Proteins , Trypanosoma , Trypanosoma/metabolism , Trypanosoma/genetics , Extracellular Vesicles/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Animals , Calcium/metabolism , Biomarkers , Trypanosomiasis/parasitology , Proteome , Epitopes/immunology
13.
Front Cell Infect Microbiol ; 14: 1455259, 2024.
Article in English | MEDLINE | ID: mdl-39228894

ABSTRACT

Introduction: Tularemia, caused by the bacterium Francisella tularensis, poses health risks to humans and can spread through a variety of routes. It has also been classified as a Tier 1 Select agent by the CDC, highlighting its potential as a bioterrorism agent. Moreover, it is difficult to diagnose in a timely fashion, owing to the non-specific nature of tularemia infections. Rapid, sensitive, and accurate detection methods are required to reduce mortality rates. We aimed to develop antibodies directed against the outer membrane protein A of F. tularensis (FopA) for rapid and accurate diagnosis of tularemia. Methods: We used a baculovirus insect cell expression vector system to produce the FopA antigen and generate anti-FopA antibodies through immunization of BALB/c mice. We then employed hybridoma and phage display technologies to screen for antibodies that could recognize unique epitopes on FopA. Result: Two monoclonal antibodies, 6B12 and 3C1, identified through phage display screening specifically bound to recombinant FopA in a dose-dependent manner. The binding affinity of the anti-FopA 6B12 and 3C1 antibodies was observed to have an equilibrium dissociation constant of 1.76 × 10-10 M and 1.32 × 10-9 M, respectively. These antibodies were used to develop a sandwich ELISA system for the diagnosis of tularemia. This assay was found to be highly specific and sensitive, with detection limits ranging from 0.062 ng/mL in PBS to 0.064 ng/mL in skim milk matrices. Discussion: Our findings demonstrate the feasibility of a novel diagnostic approach for detecting F. tularensis based on targeting FopA, as opposed to existing tests that target the bacterial lipopolysaccharide.


Subject(s)
Antibodies, Bacterial , Antibodies, Monoclonal , Bacterial Outer Membrane Proteins , Francisella tularensis , Mice, Inbred BALB C , Recombinant Proteins , Tularemia , Tularemia/diagnosis , Animals , Francisella tularensis/immunology , Francisella tularensis/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Antibodies, Monoclonal/immunology , Mice , Immunoassay/methods , Sensitivity and Specificity , Female , Cell Surface Display Techniques , Epitopes/immunology , Enzyme-Linked Immunosorbent Assay/methods , Humans , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Hybridomas , Baculoviridae/genetics
14.
Sci Rep ; 14(1): 20418, 2024 09 03.
Article in English | MEDLINE | ID: mdl-39223292

ABSTRACT

The epidemic and outbreaks of influenza B Victoria lineage (Bv) during 2019-2022 led to an analysis of genetic, epitopes, charged amino acids and Bv outbreaks. Based on the National Influenza Surveillance Network (NISN), the Bv 72 strains isolated during 2019-2022 were selected by spatio-temporal sampling, then were sequenced. Using the Compare Means, Correlate and Cluster, the outbreak data were analyzed, including the single nucleotide variant (SNV), amino acid (AA), epitope, evolutionary rate (ER), Shannon entropy value (SV), charged amino acid and outbreak. With the emergence of COVID-19, the non-pharmaceutical interventions (NPIs) made Less distant transmission and only Bv outbreak. The 2021-2022 strains in the HA genes were located in the same subset, but were distinct from the 2019-2020 strains (P < 0.001). The codon G → A transition in nucleotide was in the highest ratio but the transversion of C → A and T → A made the most significant contribution to the outbreaks, while the increase in amino acid mutations characterized by polar, acidic and basic signatures played a key role in the Bv epidemic in 2021-2022. Both ER and SV were positively correlated in HA genes (R = 0.690) and NA genes (R = 0.711), respectively, however, the number of mutations in the HA genes was 1.59 times higher than that of the NA gene (2.15/1.36) from the beginning of 2020 to 2022. The positively selective sites 174, 199, 214 and 563 in HA genes and the sites 73 and 384 in NA genes were evolutionarily selected in the 2021-2022 influenza outbreaks. Overall, the prevalent factors related to 2021-2022 influenza outbreaks included epidemic timing, Tv, Ts, Tv/Ts, P137 (B → P), P148 (B → P), P199 (P → A), P212 (P → A), P214 (H → P) and P563 (B → P). The preference of amino acid mutations for charge/pH could influence the epidemic/outbreak trends of infectious diseases. Here was a good model of the evolution of infectious disease pathogens. This study, on account of further exploration of virology, genetics, bioinformatics and outbreak information, might facilitate further understanding of their deep interaction mechanisms in the spread of infectious diseases.


Subject(s)
Disease Outbreaks , Evolution, Molecular , Influenza, Human , Mutation , Polymorphism, Single Nucleotide , Humans , Influenza, Human/epidemiology , Influenza, Human/virology , Influenza, Human/genetics , Influenza B virus/genetics , Amino Acids/genetics , Epitopes/genetics , Phylogeny , Amino Acid Substitution , Hemagglutinin Glycoproteins, Influenza Virus/genetics
15.
NPJ Syst Biol Appl ; 10(1): 101, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251627

ABSTRACT

The identification of antibody-specific epitopes on virus proteins is crucial for vaccine development and drug design. Nonetheless, traditional wet-lab approaches for the identification of epitopes are both costly and labor-intensive, underscoring the need for the development of efficient and cost-effective computational tools. Here, EpiScan, an attention-based deep learning framework for predicting antibody-specific epitopes, is presented. EpiScan adopts a multi-input and single-output strategy by designing independent blocks for different parts of antibodies, including variable heavy chain (VH), variable light chain (VL), complementary determining regions (CDRs), and framework regions (FRs). The block predictions are weighted and integrated for the prediction of potential epitopes. Using multiple experimental data samples, we show that EpiScan, which only uses antibody sequence information, can accurately map epitopes on specific antigen structures. The antibody-specific epitopes on the receptor binding domain (RBD) of SARS coronavirus 2 (SARS-CoV-2) were located by EpiScan, and the potentially valuable vaccine epitope was identified. EpiScan can expedite the epitope mapping process for high-throughput antibody sequencing data, supporting vaccine design and drug development. Availability: For the convenience of related wet-experimental researchers, the source code and web server of EpiScan are publicly available at https://github.com/gzBiomedical/EpiScan .


Subject(s)
Epitope Mapping , Epitopes , SARS-CoV-2 , Epitope Mapping/methods , SARS-CoV-2/immunology , Humans , Epitopes/immunology , Antibodies, Viral/immunology , Deep Learning , COVID-19/immunology , Computational Biology/methods , Spike Glycoprotein, Coronavirus/immunology , Software
16.
Virol J ; 21(1): 213, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252044

ABSTRACT

High-risk human papilloma virus (HR-HPV) persistent infection is closely associated with the development of cervical cancer and squamous intraepithelial lesion (SIL).The α-9 HPVs, which is predominantly composed of HR-HPV types, account for 75% of HR-HPV infection in Sichuan. The oncoproteins E6 and E7 of HPV play a crucial role in tumor initiation and progression. Notably, HPV-35 is the only HR-HPV type within the α-9 genus that is not included in the nine-valent HPV prophylactic vaccine. Cervical cell samples obtained from Sichuan were collected for HPV detection and genotyping. Among the 406 HPV-positive samples, 31 HPV-35 were detected, 24 HPV-35 E6 and 26 E7 were successfully amplified and sequenced, five nucleotide mutations in E6 and three in E7 were detected, T232C, T434G of E6 (W78R, I145R) and C67T, G84T of E7 (H23Y, L28F) were non-synonymy mutation. PAML 4.8 server was used to detect positive selection sites of HPV-35 E6, E7, and E6 is W78R. Phyre2 were used to predict and analyze protein structures, W78R made influences on protein structure. IEDB were used to screen epitopes vaccine target for HPV-35 affection therapy, and 5 HPV-35 E6 and 3 HPV-35 E7 most potential epitopes were obtained, the most potential peptides for therapy vaccine design were 79-91YRYSVYGETLEKQ, 45-60FACYDLCIVREGQPY, 124-135RFHNIGGRWTGR of E6; 3-19GEITTLQDYVLDLEPEA, 38-47TIDGPAGQAK, 70-88VQSTHIDIRKLEDLLMGTF of E7 and W78R mainly decreased the epitopes affinity.Conclusions Amino acid substitution in the positive selection sites of HPV-35 E6 and E7 genes have been found to influence protein structure and to decrease the overall affinity of antigen epitopes. This observation aligns with the evolutionary significance of positive selection site, which may confer advantages to the virus by making infected cells more challenging for the immune system to detect, thereby enhancing HPV's adaptability to the host environment. The polymorphism analysis of HPV-35 E6, E7 contributes to the enrichment of α-9 HPV data in Sichuan China, which is instrumental in improving the effectiveness of clinical detection. Furthermore, these findings provide a relevant theoretical foundation for the prevention and treatment of HPV-related diseases.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Female , China , Papillomavirus Infections/virology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/genetics , Polymorphism, Genetic , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Genotype , Adult , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/prevention & control , Epitopes/immunology , Epitopes/genetics , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Alphapapillomavirus/classification , Middle Aged , Mutation , Human Papillomavirus Viruses
17.
PLoS One ; 19(9): e0307877, 2024.
Article in English | MEDLINE | ID: mdl-39240891

ABSTRACT

Tuberculosis(TB) of the Central nervous system (CNS) is a rare and highly destructive disease. The emergence of drug resistance has increased treatment difficulty, leaving the Bacillus Calmette-Guérin (BCG) vaccine as the only licensed preventative immunization available. This study focused on identifying the epitopes of PknD (Rv0931c) and Rv0986 from Mycobacterium tuberculosis(Mtb) strain H37Rv using an in silico method. The goal was to develop a therapeutic mRNA vaccine for preventing CNS TB. The vaccine was designed to be non-allergenic, non-toxic, and highly antigenic. Codon optimization was performed to ensure effective translation in the human host. Additionally, the secondary and tertiary structures of the vaccine were predicted, and molecular docking with TLR-4 was carried out. A molecular dynamics simulation confirmed the stability of the complex. The results indicate that the vaccine structure shows effectiveness. Overall, the constructed vaccine exhibits ideal physicochemical properties, immune response, and stability, laying a theoretical foundation for future laboratory experiments.


Subject(s)
Computer Simulation , Molecular Docking Simulation , Molecular Dynamics Simulation , Mycobacterium tuberculosis , Tuberculosis, Central Nervous System , Humans , Mycobacterium tuberculosis/immunology , Tuberculosis, Central Nervous System/prevention & control , Tuberculosis, Central Nervous System/immunology , Tuberculosis Vaccines/immunology , Epitopes/immunology , Epitopes/chemistry , mRNA Vaccines , Vaccines, Synthetic/immunology
18.
Article in English | MEDLINE | ID: mdl-39258658

ABSTRACT

Rotavirus, a dsRNA virus in the Reoviridae family, shows a segmented genome. The VP1 gene encodes the RNA-dependent RNA polymerase (RdRp). This study aims to develop a multiepitope-based vaccine targeting RdRp using immunoinformatic approaches. In this study, 100 available nucleotide sequences of VP1-Rotavirus belonging to different strains across the world were retrieved from NCBI database. The selected sequences were aligned, and a global consensus sequence was developed by using CLC work bench. The study involved immunoinformatic approaches and molecular docking studies to reveal the promiscuous epitopes that can be eventually used as active vaccine candidates for Rotavirus. In total, 27 highly immunogenic, antigenic, and non-allergenic T-cell and B-cell epitopes were predicted for the Multiepitope vaccine (MEV) against rotavirus. It was also observed that MEV can prove to be effective worldwide due to its high population coverage, demonstrating the consistency of this vaccine. Moreover, there is a high docking interaction and immunological response with a binding score of -50.2 kcal/mol, suggesting the vaccine's efficacy. Toll-like receptors (TLRs) also suggest that the vaccine is physiologically and immunologically effective. Collectively, our data point to an effective MEV against rotavirus that can effectively reduce viral infections and improve the health status worldwide.


Subject(s)
Molecular Docking Simulation , Rotavirus Vaccines , Rotavirus , Vaccines, Subunit , Rotavirus/immunology , Rotavirus/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Rotavirus Vaccines/immunology , RNA-Dependent RNA Polymerase/immunology , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/chemistry , Computational Biology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Humans , Epitopes/immunology , Epitopes/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Rotavirus Infections/prevention & control , Rotavirus Infections/immunology , Immunoinformatics , Protein Subunit Vaccines
19.
Nutrients ; 16(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39275185

ABSTRACT

BACKGROUND: Cross-reactivity between nonspecific lipid transfer proteins could cause anaphylaxis, further influencing food avoidance and nutrient deficiencies. The one affecting olive pollen (Ole e 7) and peach (Pru p 3) may underlie a variety of pollen-food syndromes, though a deep molecular analysis is necessary. METHODS: Three Ole e 7-monosensitised patients (MON_OLE), three Pru p 3-monosensitised patients (MON_PRU) and three bisensitised patients (BI) were selected. For epitope mapping, both digested proteins were incubated with patient sera, and the captured IgE-bound peptides were characterised by LC-MS. RESULTS: The analysis revealed two Ole e 7 epitopes and the three Pru p 3 epitopes previously described. Interestingly, the "KSALALVGNKV" Ole e 7 peptide was recognised by MON_OLE, BI and MON_PRU patients. Conversely, all patients recognised the "ISASTNCATVK" Pru p 3 peptide. Although complete sequence alignment between both proteins revealed 32.6% identity, local alignment considering seven residue fragments showed 50 and 57% identity when comparing "ISASTNCATVK" with Ole e 7 and "KSALALVGNKV" with Pru p 3. CONCLUSIONS: This study mapped sIgE-Ole e 7-binding epitopes, paving the way for more precise diagnostic tools. Assuming non-significant sequence similarity, structural homology and shared key residues may underlie the potential cross-reactivity between Ole e 7 and Pru p 3 nsLTPs.


Subject(s)
Antigens, Plant , Cross Reactions , Food Hypersensitivity , Immunoglobulin E , Olea , Plant Proteins , Pollen , Prunus persica , Humans , Antigens, Plant/immunology , Food Hypersensitivity/immunology , Pollen/immunology , Immunoglobulin E/immunology , Immunoglobulin E/blood , Olea/immunology , Plant Proteins/immunology , Female , Male , Prunus persica/immunology , Epitope Mapping , Adult , Rhinitis, Allergic, Seasonal/immunology , Amino Acid Sequence , Epitopes/immunology , Allergens/immunology , Middle Aged , Carrier Proteins/immunology
20.
Curr Microbiol ; 81(11): 366, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39297982

ABSTRACT

In post-COVID-19 syndrome, clinical presentation of the nerve fiber dysfunction plays an important role. The possibility of autoantigen cross-mimicry of human coronaviruses and the peripheral nervous system needs to be investigated. The bioinformatic analysis was applied to search for possible common protein sequences located in the immunoreactive epitopes. Among the autoantigens of the human nervous system, fibroblast growth factor receptor protein 3, myelin protein P0, myelin protein P2, sodium channel protein type 9, alpha protein subunit, plexin-D1 protein and ubiquitin-carboxyl-terminal hydrolase protein of the L1 isoenzyme were selected. The original "Alignmentaj" analytical program was created. The UniProt database, Protein Data Bank, and AlphaFold databases were used. The analysis of protein sequence similarities of spike glycoproteins in human coronaviruses revealed common pentapeptides of the MERS-CoV-2 virus with the fibroblast growth factor receptor 3 and myelin protein P2. Among seasonal coronaviruses, common peptide sequences were identified in HCoV-HKU-1 virus with sodium channel protein type 9 subunit alpha and Plexin-D1, HCoV-OC43 with Plexin-D1, as well as HCoV-NL63 with Plexin-D1 and Ubiquitin carboxyl-terminal hydrolase isozyme L1. Some shared peptides belong to immunoreactive epitopes. The most important targets for the molecular similarities are the sodium channel subunits and fibroblast growth factor receptor 3, both for seasonal and highly pathogenic coronaviruses. The data obtained make it possible to identify new potential targets for the development of autoimmune reactions that may occur against the background of the infections with highly pathogenic as well as seasonal coronaviruses.


Subject(s)
Autoantigens , Molecular Mimicry , Humans , Autoantigens/immunology , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antigens, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Epitopes/immunology , Computational Biology , Coronavirus/immunology , Coronavirus/genetics , Amino Acid Sequence
SELECTION OF CITATIONS
SEARCH DETAIL