Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.556
Filter
1.
Gut Microbes ; 16(1): 2350150, 2024.
Article in English | MEDLINE | ID: mdl-38841888

ABSTRACT

Comensal Bacteroidota (Bacteroidota) and Enterobacteriacea are often linked to gut inflammation. However, the causes for variability of pro-inflammatory surface antigens that affect gut commensal/opportunistic dualism in Bacteroidota remain unclear. By using the classical lipopolysaccharide/O-antigen 'rfb operon' in Enterobacteriaceae as a surface antigen model (5-rfb-gene-cluster rfbABCDX), and a recent rfbA-typing strategy for strain classification, we characterized the integrity and conservancy of the entire rfb operon in Bacteroidota. Through exploratory analysis of complete genomes and metagenomes, we discovered that most Bacteroidota have the rfb operon fragmented into nonrandom patterns of gene-singlets and doublets/triplets, termed 'rfb-gene-clusters', or rfb-'minioperons' if predicted as transcriptional. To reflect global operon integrity, contiguity, duplication, and fragmentation principles, we propose a six-category (infra/supra-numerary) cataloging system and a Global Operon Profiling System for bacteria. Mechanistically, genomic sequence analyses revealed that operon fragmentation is driven by intra-operon insertions of predominantly Bacteroides-DNA (thetaiotaomicron/fragilis) and likely natural selection in gut-wall specific micro-niches or micropathologies. Bacteroides-insertions, also detected in other antigenic operons (fimbriae), but not in operons deemed essential (ribosomal), could explain why Bacteroidota have fewer KEGG-pathways despite large genomes. DNA insertions, overrepresenting DNA-exchange-avid (Bacteroides) species, impact our interpretation of functional metagenomics data by inflating by inflating gene-based pathway inference and by overestimating 'extra-species' abundance. Of disease relevance, Bacteroidota species isolated from cavitating/cavernous fistulous tract (CavFT) microlesions in Crohn's Disease have supra-numerary fragmented operons, stimulate TNF-alpha from macrophages with low potency, and do not induce hyperacute peritonitis in mice compared to CavFT Enterobacteriaceae. The impact of 'foreign-DNA' insertions on pro-inflammatory operons, metagenomics, and commensalism/opportunism requires further studies to elucidate their potential for novel diagnostics and therapeutics, and to elucidate the role of co-existing pathobionts in Crohn's disease microlesions.


Subject(s)
Crohn Disease , Gastrointestinal Microbiome , Metagenomics , Operon , Mice , Animals , Humans , Crohn Disease/microbiology , Crohn Disease/genetics , Bacteroidetes/genetics , Bacteroidetes/classification , Antigens, Bacterial/genetics , Genome, Bacterial , Enterobacteriaceae/genetics , Enterobacteriaceae/classification
2.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38834194

ABSTRACT

Vinculin is a cytoskeletal linker strengthening cell adhesion. The Shigella IpaA invasion effector binds to vinculin to promote vinculin supra-activation associated with head-domain-mediated oligomerization. Our study investigates the impact of mutations of vinculin D1D2 subdomains' residues predicted to interact with IpaA VBS3. These mutations affected the rate of D1D2 trimer formation with distinct effects on monomer disappearance, consistent with structural modeling of a closed and open D1D2 conformer induced by IpaA. Notably, mutations targeting the closed D1D2 conformer significantly reduced Shigella invasion of host cells as opposed to mutations targeting the open D1D2 conformer and later stages of vinculin head-domain oligomerization. In contrast, all mutations affected the formation of focal adhesions (FAs), supporting the involvement of vinculin supra-activation in this process. Our findings suggest that IpaA-induced vinculin supra-activation primarily reinforces matrix adhesion in infected cells, rather than promoting bacterial invasion. Consistently, shear stress studies pointed to a key role for IpaA-induced vinculin supra-activation in accelerating and strengthening cell-matrix adhesion.


Subject(s)
Cell Adhesion , Focal Adhesions , Vinculin , Vinculin/metabolism , Vinculin/genetics , Humans , Focal Adhesions/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Mutation , Host-Pathogen Interactions , HeLa Cells , Protein Binding , Shigella/metabolism , Shigella/genetics , Antigens, Bacterial/metabolism , Antigens, Bacterial/genetics , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/metabolism
3.
Arq Gastroenterol ; 61: e23139, 2024.
Article in English | MEDLINE | ID: mdl-38775582

ABSTRACT

BACKGROUND: Helicobacter pylori (H. pylori) is a gram-negative bacterium associated with the etiology of several gastrointestinal tract pathologies, and cagA-positive (cagA+) strains are found in populations with gastric ulcers and precancerous lesions, inducing pro-inflammatory responses. The development of neoplasms is related to microRNA (miRNA) dysregulation, indicating highly expressed miRNA-629. The article aims to correlate the expression level of miRNA-629 with the presence of H. pylori and the pathogenicity marker cagA. METHODS: 203 gastric biopsy samples were evaluated from individuals with normal gastric tissue (n=60), gastritis (n=96), and gastric cancer (n=47) of both genders and over 18 years old. The samples were subdivided according to the presence or absence of H. pylori, detected by polymerase chain reaction (PCR). RNA was extracted using a commercial kit and quantified. Complementary DNA (cDNA) was synthesized using commercial kits, and the relative expression was calculated using the 2-ΔΔCt method. RESULTS: Individuals infected with H. pylori are nine times more likely to develop gastric cancer. Cancer patients appeared to have decreased expression of miRNA-629; however, the presence of the bacterium would not influence this reduction. Individuals in the cancer group showed lower miRNA-629 expression when cagA+; however, in the control group, the expression was higher when cagA+. CONCLUSION: H. pylori is a factor involved in the etiology and progression of gastric diseases. Reduction in miRNA-629 expression in cancer patients occurs independent of the presence of the bacterium, but when the cagA pathogenicity marker is present, it induces changes in the gene expression of the respective miRNA.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Helicobacter Infections , Helicobacter pylori , MicroRNAs , Stomach Neoplasms , Humans , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Stomach Neoplasms/microbiology , Stomach Neoplasms/genetics , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , MicroRNAs/genetics , MicroRNAs/analysis , Female , Male , Helicobacter Infections/microbiology , Middle Aged , Adult , Aged , Gastritis/microbiology
4.
Front Immunol ; 15: 1392456, 2024.
Article in English | MEDLINE | ID: mdl-38779673

ABSTRACT

In response to the global threat posed by bacterial pathogens, which are the second leading cause of death worldwide, vaccine development is challenged by the diversity of bacterial serotypes and the lack of immunoprotection across serotypes. To address this, we introduce BacScan, a novel genome-wide technology for the rapid discovery of conserved highly immunogenic proteins (HIPs) across serotypes. Using bacterial-specific serum, BacScan combines phage display, immunoprecipitation, and next-generation sequencing to comprehensively identify all the HIPs in a single assay, thereby paving the way for the development of universally protective vaccines. Our validation of this technique with Streptococcus suis, a major pathogenic threat, led to the identification of 19 HIPs, eight of which conferred 20-100% protection against S. suis challenge in animal models. Remarkably, HIP 8455 induced complete immunity, making it an exemplary vaccine target. BacScan's adaptability to any bacterial pathogen positions it as a revolutionary tool that can expedite the development of vaccines with broad efficacy, thus playing a critical role in curbing bacterial transmission and slowing the march of antimicrobial resistance.


Subject(s)
Bacterial Proteins , Animals , Mice , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Streptococcal Infections/immunology , Streptococcal Infections/prevention & control , Streptococcus suis/immunology , Streptococcus suis/genetics , Genome, Bacterial , High-Throughput Nucleotide Sequencing , Female , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Humans , Bacterial Vaccines/immunology
5.
Cell Mol Biol Lett ; 29(1): 70, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741147

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis heat-resistant antigen (Mtb-HAg) is a peptide antigen released from the mycobacterial cytoplasm into the supernatant of Mycobacterium tuberculosis (Mtb) attenuated H37Ra strain after autoclaving at 121 °C for 20 min. Mtb-HAg can specifically induce γδ T-cell proliferation in vitro. However, the exact composition of Mtb-HAg and the protein antigens that are responsible for its function are currently unknown. METHODS: Mtb-HAg extracted from the Mtb H37Ra strain was subjected to LC‒MS mass spectrometry. Twelve of the identified protein fractions were recombinantly expressed in Escherichia coli by genetic engineering technology using pET-28a as a plasmid and purified by Ni-NTA agarose resin to stimulate peripheral blood mononuclear cells (PBMCs) from different healthy individuals. The proliferation of γδ T cells and major γδ T-cell subset types as well as the production of TNF-α and IFN-γ were determined by flow cytometry. Their proliferating γδ T cells were isolated and purified using MACS separation columns, and Mtb H37Ra-infected THP-1 was co-cultured with isolated and purified γδ T cells to quantify Mycobacterium viability by counting CFUs. RESULTS: In this study, Mtb-HAg from the attenuated Mtb H37Ra strain was analysed by LC‒MS mass spectrometry, and a total of 564 proteins were identified. Analysis of the identified protein fractions revealed that the major protein components included heat shock proteins and Mtb-specific antigenic proteins. Recombinant expression of 10 of these proteins in by Escherichia coli genetic engineering technology was used to successfully stimulate PBMCs from different healthy individuals, but 2 of the proteins, EsxJ and EsxA, were not expressed. Flow cytometry results showed that, compared with the IL-2 control, HspX, GroEL1, and GroES specifically induced γδ T-cell expansion, with Vγ2δ2 T cells as the main subset, and the secretion of the antimicrobial cytokines TNF-α and IFN-γ. In contrast, HtpG, DnaK, GroEL2, HbhA, Mpt63, EsxB, and EsxN were unable to promote γδ T-cell proliferation and the secretion of TNF-α and IFN-γ. None of the above recombinant proteins were able to induce the secretion of TNF-α and IFN-γ by αß T cells. In addition, TNF-α, IFN-γ-producing γδ T cells inhibit the growth of intracellular Mtb. CONCLUSION: Activated γδ T cells induced by Mtb-HAg components HspX, GroES, GroEL1 to produce TNF-α, IFN-γ modulate macrophages to inhibit intracellular Mtb growth. These data lay the foundation for subsequent studies on the mechanism by which Mtb-HAg induces γδ T-cell proliferation in vitro, as well as the development of preventive and therapeutic vaccines and rapid diagnostic reagents.


Subject(s)
Antigens, Bacterial , Cell Proliferation , Mycobacterium tuberculosis , T-Lymphocytes , Humans , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Antigens, Bacterial/genetics , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Tumor Necrosis Factor-alpha/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology
6.
Curr Microbiol ; 81(7): 197, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816607

ABSTRACT

Identifying and evaluating potential vaccine candidates has become one of the main objectives to combat tuberculosis. Among them, mannosylated Apa antigen from Mycobacterium tuberculosis and the non-mannosylated protein expressed in Escherichia coli, have been studied. Although both proteins can induce a protective response in mice, it has been considered that native protein can be dispensed. In this work, we study the protective response induced by Apa expressed in E. coli and in Streptomyces lividans. The latter, like native is secreted as a double band of 45/47 kDa, however, only its 47 kDa band is mannosylated. Both antigens and BCG were intranasal administrated in mice, and animals were then challenged by aerosol with M. tuberculosis H37Rv. The results showed that both, Apa from S. lividans and E. coli conferred statistically significantly protection to animals compared to controls. The cytokine immune response was studied by an immunoassay after animals' immunization, revealing that Apa from S. lividans induced a statistically significant proliferation of T cell, as well as the expression of IFN-γ, IL-1ß, IL-17 and IL-10. In contrast, non-proliferation was obtained with non-mannosylated protein, but induction of IL-12 and IL-17 was observed. Together, these results demonstrate that both proteins were able to modulate a specific immune response against M. tuberculosis, that could be driven by different mechanisms possibly associated with the presence or not of mannosylation. Furthermore, stimulation of cells from BCG-vaccinated animals with the proteins could be an important tool, to help define the use of a given subunit-vaccine after BCG vaccination.


Subject(s)
Administration, Intranasal , Cytokines , Mycobacterium tuberculosis , Streptomyces lividans , Tuberculosis , Animals , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/genetics , Mice , Cytokines/metabolism , Tuberculosis/prevention & control , Tuberculosis/immunology , Streptomyces lividans/genetics , Streptomyces lividans/immunology , Aerosols , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/administration & dosage , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Mice, Inbred BALB C , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/administration & dosage
7.
Diagn Microbiol Infect Dis ; 109(3): 116338, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38718661

ABSTRACT

The diagnosis if leprosy is difficult, as it requires clinical expertise and sensitive laboratory tests. In this study, we develop a serological test for leprosy by using bioinformatics tools to identify specific B-cell epitopes from Mycobacterium leprae hypothetical proteins, which were used to construct a recombinant chimeric protein, M1. The synthetic peptides were obtained and showed good reactivity to detect leprosy patients, although the M1 chimera have showed sensitivity (Se) and specificity (Sp) values higher than 90.0% to diagnose both paucibacillary (PB) and multibacillary (MB) leprosy patients, but not those developing tegumentary or visceral leishmaniasis, tuberculosis, Chagas disease, malaria, histoplasmosis and aspergillosis, in ELISA experiments. Using sera from household contacts, values for Se and Sp were 100% and 65.3%, respectively. In conclusion, our proof-of-concept study has generated data that suggest that a new recombinant protein could be developed into a diagnostic antigen for leprosy.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Epitopes, B-Lymphocyte , Leprosy , Mycobacterium leprae , Sensitivity and Specificity , Humans , Mycobacterium leprae/immunology , Mycobacterium leprae/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Leprosy/diagnosis , Leprosy/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Enzyme-Linked Immunosorbent Assay/methods , Adult , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Male , Female , Serologic Tests/methods , Computational Biology/methods , Middle Aged , Young Adult , Adolescent
8.
Int Immunopharmacol ; 134: 112160, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38710117

ABSTRACT

INTRODUCTION: Cholera is a severe gastrointestinal disease that manifests with rapid onset of diarrhea, vomiting, and high mortality rates. Due to its widespread occurrence in impoverished communities with poor water sanitation, there is an urgent demand for a cost-effective and highly efficient vaccine. Multi-epitope vaccines containing dominant immunological epitopes and adjuvant compounds have demonstrated potential in boosting the immune response. MATERIAL AND METHODS: B and T epitopes of OMPU, OMPW, TCPA, CTXA, and CTXB proteins were predicted using bioinformatics methods. Subsequently, highly antigenic multi-epitopes that are non-allergenic and non-toxic were synthesized. These multi-epitopes were then cloned into the pCOMB phagemid. A plasmid M13KO7ΔpIII containing all helper phage proteins except pIII was created to produce the recombinant phage. Female Balb/c mice were divided into three groups and immunized accordingly. The mice received the helper phage, recombinant phage or PBS via gavage feeding thrice within two weeks. Serum samples were collected before and after immunization for the ELISA test as well as evaluating immune system induction through ELISpot testing of spleen lymphocytes. RESULTS: The titer of the recombinant phage was determined to be 1011 PFU/ml. The presence of the recombinant phage was confirmed through differences in optical density between sample and control groups in the ELISA phage technique, as well as by observing transduction activity, which demonstrated successful production of a recombinant phage displaying the Vibrio multi-epitope on M13 phage pIII. ELISA results revealed significant differences in phage antibodies before and after inoculation, particularly notable in the negative control mice. Mice treated with multi-epitope phages exhibited antibodies against Vibrio cholerae lysate. Additionally, ELISpot results indicated activation of cellular immunity in mice receiving both Vibrio and helper phage. CONCLUSION: This study emphasizes the potential of multi-epitope on phage to enhance both cellular and humoral immunity in mice, demonstrating how phages can be used as adjuvants to stimulate mucosal immunity and act as promising candidates for oral vaccination.


Subject(s)
Antibodies, Bacterial , Cholera Vaccines , Cholera , Immunity, Cellular , Immunity, Humoral , Mice, Inbred BALB C , Vibrio cholerae , Animals , Vibrio cholerae/immunology , Female , Cholera/prevention & control , Cholera/immunology , Cholera Vaccines/immunology , Cholera Vaccines/administration & dosage , Administration, Oral , Mice , Antibodies, Bacterial/blood , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Immunization , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Humans , Bacteriophages/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics
9.
Int Immunopharmacol ; 135: 112287, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38776850

ABSTRACT

Achromobacter xylosoxidans is an aerobic, catalase-positive, non-pigment-forming, Gram-negative, and motile bacterium. It potentially causes a wide range of human infections in cystic fibrosis and non-cystic fibrosis patients. However, developing a safe preventive or therapeutic solution against A. xylosoxidans remains challenging. This study aimed to construct an epitope-based vaccine candidate using immunoinformatic techniques. A. xylosoxidans was isolated from an auto workshop in Lahore, and its identification was confirmed through 16S rRNA amplification and bioinformatic analysis. Two protein targets with GenBank accession numbers AKP90890.1 and AKP90355.1 were selected for the vaccine construct. Both proteins exhibited antigenicity, with scores of 0.757 and 0.580, respectively and the epitopes were selected based on the IC50 value using the ANN 4.0 and NN-align 2.3 epitope prediction method for MHC I and MHC II epitopes respectively and predicted epitopes were analyzed for antigenicity, allergenicity and pathogenicity. The vaccine construct demonstrated structural stability, thermostability, solubility, and hydrophilicity. The vaccine produced 250 B-memory cells per mm3 and approximately 16,000 IgM + IgG counts, indicating an effective immune response against A. xylosoxidans. Moreover, the vaccine candidate interacted stably with toll-like receptor 5, a pattern recognition receptor, with a confidence score of 0.98. These results highlight the potency of the designed vaccine candidate, suggesting its potential to withstand rigorous in vitro and in vivo clinical trials. This epitope-based vaccine could serve as the first preventive immunotherapy against A. xylosoxidans infections, addressing this bacterium's health and financial burdens. The findings demonstrate the value of employing immunoinformatic tools in vaccine development, paving the way for more precise and tailored approaches to combating microbial threats.


Subject(s)
Achromobacter denitrificans , Bacterial Vaccines , Gram-Negative Bacterial Infections , RNA, Ribosomal, 16S , Achromobacter denitrificans/immunology , Achromobacter denitrificans/genetics , Bacterial Vaccines/immunology , Humans , RNA, Ribosomal, 16S/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/microbiology , Animals , Epitopes/immunology , Computer Simulation , Female , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Mice , Computational Biology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics
10.
Gene ; 920: 148526, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38703866

ABSTRACT

INTRODUCTION: Outer membrane protein (OMP) of Helicobacter pylori (H. pylori) i.e., blood group antigen binding adhesin (babA) is responsible for the attachment of H. pylori in the gastric epithelium. Its adherence is causative for gastric pathology such as gastritis, peptic ulcer disease (PUD), or digestive tract disorders like erosive reflux disease (ERD) and (NERD) non-erosive reflux disease and together called Gastroesophageal reflux disease (GERD). BabA manifests rapid and varied selection via substitution of amino acid in its Leb-carbohydrate binding domain (CBD) which enables better binding preferences for distinct human populations and ABO blood group phenotypes. The positive evolutionary selection of the pathogenic factor of this genetically diverse bacterium has enabled it to adapt to the host gastric environment. Analyzing the association of virulent genes (cagA, vacA) and babA will help us better understand bacteria's pathogenicity. METHOD: 109 H. pylori strains from patients with distinct gastrointestinal diseases were genotyped using Polymerase Chain Reaction(PCR) for cagA, vacA, and babA followed by Sanger sequencing and phylogenetic analysis. RESULT: In the babA + ve genotype, a statistically significant association with p = 0.04 and < 0.0001 is seen in gastritis and ERD respectively. A significant association of genotype vacAs1m2 (p = 0.0002) was seen in gastritis, vacAs1m1 (p = 0.02) in NERD, vacAs1m1 (p < 0.0001) and vacAs1m2 (p = 0.002) in ERD. This relationship helps to detect gastritis or ERD where BabA gene can be used as an independent marker for detecting their presence. CONCLUSION: The appearance of variants within distinct disease categories is due to local genetic variation.


Subject(s)
Adhesins, Bacterial , Helicobacter Infections , Helicobacter pylori , Phylogeny , Humans , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Helicobacter pylori/isolation & purification , Adhesins, Bacterial/genetics , Helicobacter Infections/microbiology , India , Male , Gastritis/microbiology , Female , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/genetics , Antigens, Bacterial/genetics , Genotype , Adult , Middle Aged , Bacterial Proteins/genetics
11.
Biosens Bioelectron ; 256: 116282, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38626615

ABSTRACT

Helicobacter pylori (H. pylori) infection correlates closely with gastric diseases such as gastritis, ulcers, and cancer, influencing more than half of the world's population. Establishing a rapid, precise, and automated platform for H. pylori diagnosis is an urgent clinical need and would significantly benefit therapeutic intervention. Recombinase polymerase amplification (RPA)-CRISPR recently emerged as a promising molecular diagnostic assay due to its rapid detection capability, high specificity, and mild reaction conditions. In this work, we adapted the RPA-CRISPR assay on a digital microfluidics (DMF) system for automated H. pylori detection and genotyping. The system can achieve multi-target parallel detection of H. pylori nucleotide conservative genes (ureB) and virulence genes (cagA and vacA) across different samples within 30 min, exhibiting a detection limit of 10 copies/rxn and no false positives. We further conducted tests on 80 clinical saliva samples and compared the results with those derived from real-time quantitative polymerase chain reaction, demonstrating 100% diagnostic sensitivity and specificity for the RPA-CRISPR/DMF method. By automating the assay process on a single chip, the DMF system can significantly reduce the usage of reagents and samples, minimize the cross-contamination effect, and shorten the reaction time, with the additional benefit of losing the chance of experiment failure/inconsistency due to manual operations. The DMF system together with the RPA-CRISPR assay can be used for early detection and genotyping of H. pylori with high sensitivity and specificity, and has the potential to become a universal molecular diagnostic platform.


Subject(s)
Biosensing Techniques , Genotyping Techniques , Helicobacter Infections , Helicobacter pylori , Helicobacter pylori/genetics , Helicobacter pylori/isolation & purification , Humans , Helicobacter Infections/diagnosis , Helicobacter Infections/microbiology , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Genotyping Techniques/instrumentation , Genotyping Techniques/methods , Genotype , Bacterial Proteins/genetics , Nucleic Acid Amplification Techniques/methods , Nucleic Acid Amplification Techniques/instrumentation , Microfluidics/methods , Antigens, Bacterial/genetics , Antigens, Bacterial/analysis , DNA, Bacterial/genetics , DNA, Bacterial/analysis , DNA, Bacterial/isolation & purification , Recombinases/metabolism
12.
Curr Microbiol ; 81(5): 127, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575759

ABSTRACT

An urgent need is to introduce an effective vaccine against Mycobacterium tuberculosis (M.tb) infection. In the present study, a multi-stage M.tb immunodominant Fcγ1 fusion protein (Ag85B:HspX:hFcγ1) was designed and produced, and the immunogenicity of purified protein was evaluated. This recombinant fusion protein was produced in the Pichia pastoris expression system. The HiTrap-rPA column affinity chromatography purified and confirmed the fusion protein using ELISA and Western blotting methods. The co-localisation assay was used to confirm its proper folding and function. IFN-γ, IL-12, IL-4, and TGF-ß expression in C57BL/6 mice then evaluated the immunogenicity of the construct in the presence and absence of BCG. After expression optimisation, medium-scale production and the Western blotting test confirmed suitable production of Ag85B:HspX:hFcγ1. The co-localisation results on antigen-presenting cells (APCs) showed that Ag85B:HspX:hFcγ1 properly folded and bound to hFcγRI. This strong co-localisation with its receptor can confirm inducing proper Th1 responses. The in vivo immunisation assay showed no difference in the expression of IL-4 but a substantial increase in the expression of IFN-γ and IL-12 (P ≤ 0.02) and a moderate increase in TGF-ß (P = 0.05). In vivo immunisation assay revealed that Th1-inducing pathways have been stimulated, as IFN-γ and IL-12 strongly, and TGF-ß expression moderately increased in Ag85B:HspX:hFcγ1 group and Ag85B:HspX:hFcγ1+BCG. Furthermore, the production of IFN-γ from splenocytes in the Ag85B:HspX:hFcγ1 group was enormously higher than in other treatments. Therefore, this Fc fusion protein can make a selective multi-stage delivery system for inducing appropriate Th1 responses and is used as a subunit vaccine alone or in combination with others.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis Vaccines , Mice , Animals , Mycobacterium tuberculosis/genetics , Bacterial Proteins/genetics , Antigens, Bacterial/genetics , BCG Vaccine , Interleukin-4 , Mice, Inbred C57BL , Recombinant Proteins/genetics , Interleukin-12 , Transforming Growth Factor beta , Tuberculosis Vaccines/genetics , Acyltransferases/genetics
13.
mBio ; 15(5): e0069324, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38587426

ABSTRACT

Among genes present in all group A streptococci (GAS), those encoding M-fibril and T-pilus proteins display the highest levels of sequence diversity, giving rise to the two primary serological typing schemes historically used to define strain. A new genotyping scheme for the pilin adhesin and backbone genes is developed and, when combined with emm typing, provides an account of the global GAS strain population. Cluster analysis based on nucleotide sequence similarity assigns most T-serotypes to discrete pilin backbone sequence clusters, yet the established T-types correspond to only half the clusters. The major pilin adhesin and backbone sequence clusters yield 98 unique combinations, defined as "pilin types." Numerous horizontal transfer events that involve pilin or emm genes generate extensive antigenic and functional diversity on the bacterial cell surface and lead to the emergence of new strains. Inferred pilin genotypes applied to a meta-analysis of global population-based collections of pharyngitis and impetigo isolates reveal highly significant associations between pilin genotypes and GAS infection at distinct ecological niches, consistent with a role for pilin gene products in adaptive evolution. Integration of emm and pilin typing into open-access online tools (pubmlst.org) ensures broad utility for end-users wanting to determine the architecture of M-fibril and T-pilus genes from genome assemblies.IMPORTANCEPrecision in defining the variant forms of infectious agents is critical to understanding their population biology and the epidemiology of associated diseases. Group A Streptococcus (GAS) is a global pathogen that causes a wide range of diseases and displays a highly diverse cell surface due to the antigenic heterogeneity of M-fibril and T-pilus proteins which also act as virulence factors of varied functions. emm genotyping is well-established and highly utilized, but there is no counterpart for pilin genes. A global GAS collection provides the basis for a comprehensive pilin typing scheme, and online tools for determining emm and pilin genotypes are developed. Application of these tools reveals the expansion of structural-functional diversity among GAS via horizontal gene transfer, as evidenced by unique combinations of surface protein genes. Pilin and emm genotype correlations with superficial throat vs skin infection provide new insights on the molecular determinants underlying key ecological and epidemiological trends.


Subject(s)
Genetic Variation , Genotype , Streptococcus pyogenes , Streptococcus pyogenes/genetics , Streptococcus pyogenes/classification , Humans , Recombination, Genetic , Bacterial Outer Membrane Proteins/genetics , Fimbriae Proteins/genetics , Gene Transfer, Horizontal , Antigens, Bacterial/genetics , Streptococcal Infections/microbiology , Streptococcal Infections/epidemiology , Impetigo/microbiology , Impetigo/epidemiology , Pharyngitis/microbiology , Fimbriae, Bacterial/genetics , Carrier Proteins
14.
Infect Immun ; 92(5): e0044023, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38591882

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli Vaccines , Extraintestinal Pathogenic Escherichia coli , Hemolysin Proteins , Animals , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Mice , Hemolysin Proteins/immunology , Hemolysin Proteins/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Escherichia coli Vaccines/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Female , Virulence Factors/genetics , Virulence Factors/immunology , Type V Secretion Systems/immunology , Type V Secretion Systems/genetics , Disease Models, Animal , Humans
15.
Diagn Microbiol Infect Dis ; 109(3): 116299, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38642546

ABSTRACT

OBJECTIVE: To evaluate the serological diagnosis value of recombinant protein antigen Tp0608 for syphilis. METHOD: 406 patients with various stages of syphilis were enrolled. A recombinant protein antigen Tp0608 was established and ELISA was used to detect patients with various stages of syphilis. The results were compared with the conventional rapid plasma reagin test (RPR) and Treponema pallidum particle agglutination test (TPPA). The sensitivity of Tp0608 recombinant protein and RPR+TPPA screening was 96.6 % and 93.1 % respectively for patients with various stages of syphilis. For patients who may have cross reactivity, the specificity of Tp0608 recombinant protein screening is 98.9 %, and the AUC of the ROC curve is 0.99; The specificity of RPR+TPPA screening was 97.3 %, and the AUC of the ROC curve was 0.96. The sensitivity and specificity of Tp0608 recombinant protein in syphilis screening are higher than conventional RPR+TPPA methods, especially in congenital syphilis and primary syphilis. CONCLUSION: The Tp0608 recombinant protein is a promising diagnostic antigen for syphilis screening, but its intracellular location and protective response have not been determined, and further verification is needed.


Subject(s)
Antigens, Bacterial , Enzyme-Linked Immunosorbent Assay , Recombinant Proteins , Sensitivity and Specificity , Syphilis Serodiagnosis , Syphilis , Treponema pallidum , Humans , Syphilis/diagnosis , Syphilis/blood , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Treponema pallidum/immunology , Treponema pallidum/genetics , Syphilis Serodiagnosis/methods , Adult , Female , Male , Enzyme-Linked Immunosorbent Assay/methods , Middle Aged , Antibodies, Bacterial/blood , Young Adult , ROC Curve , Adolescent , Bacterial Proteins/immunology , Bacterial Proteins/genetics
16.
Genes (Basel) ; 15(4)2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38674361

ABSTRACT

Bacillus anthracis is the bacterium responsible for causing the zoonotic disease called anthrax. The disease presents itself in different forms like gastrointestinal, inhalation, and cutaneous. Bacterial spores are tremendously adaptable, can persist for extended periods and occasionally endanger human health. The Anthrax Toxin Receptor-2 (ANTXR2) gene acts as membrane receptor and facilitates the entry of the anthrax toxin into host cells. Additionally, mutations in the ANTXR2 gene have been linked to various autoimmune diseases, including Hyaline Fibromatosis Syndrome (HFS), Ankylosing Spondylitis (AS), Juvenile Hyaline Fibromatosis (JHF), and Infantile Systemic Hyalinosis (ISH). This study delves into the genetic landscape of ANTXR2, aiming to comprehend its associations with diverse disorders, elucidate the impacts of its mutations, and pinpoint minimal non-pathogenic mutations capable of reducing the binding affinity of the ANTXR2 gene with the protective antigen. Recognizing the pivotal role of single-nucleotide polymorphisms (SNPs) in shaping genetic diversity, we conducted computational analyses to discern highly deleterious and tolerated non-synonymous SNPs (nsSNPs) in the ANTXR2 gene. The Mutpred2 server determined that the Arg465Trp alteration in the ANTXR2 gene leads to altered DNA binding (p = 0.22) with a probability of a deleterious mutation of 0.808; notably, among the identified deleterious SNPs, rs368288611 (Arg465Trp) stands out due to its significant impact on altering the DNA-binding ability of ANTXR2. We propose these SNPs as potential candidates for hypertension linked to the ANTXR2 gene, which is implicated in blood pressure regulation. Noteworthy among the tolerated substitutions is rs200536829 (Ala33Ser), recognized as less pathogenic; this highlights its potential as a valuable biomarker, potentially reducing side effects on the host while also reducing binding with the protective antigen protein. Investigating these SNPs holds the potential to correlate with several autoimmune disorders and mitigate the impact of anthrax disease in humans.


Subject(s)
Anthrax , Antigens, Bacterial , Mutation , Polymorphism, Single Nucleotide , Receptors, Peptide , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Humans , Anthrax/microbiology , Anthrax/genetics , Anthrax/immunology , Receptors, Peptide/genetics , Bacterial Toxins/genetics , Bacillus anthracis/genetics , Bacillus anthracis/pathogenicity , Hyaline Fibromatosis Syndrome/genetics , Hyaline Fibromatosis Syndrome/microbiology , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/microbiology , Disease Resistance/genetics , Receptors, Cell Surface/genetics , Protein Binding
17.
Braz J Microbiol ; 55(2): 1393-1404, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38676790

ABSTRACT

Helicobacter pylori is the most common cause of gastroduodenal diseases. The concept that cagA-positive H. pylori is a risk factor for gastric cancer appears to be true only for H. pylori strains from Western countries. Other virulent genes may have a synergistic interaction with cagA during pathogenesis. This study aims to investigate H. pylori cagA, vacA, and iceA prevalence, genotypes, and their association to clinical outcomes in Vietnamese patients. The cagA status and vacA and iceA genotypes were determined using the PCR technique on DNA extracted from gastric biopsies of 141 patients with gastroduodenal diseases. After performing molecular analysis for cagA, vacA, and iceA genes, samples with mixed H. pylori strains, positivity, or negativity for both cagA and cagPAI-empty site, or unidentified genotypes were excluded. Finally, 107 samples were examined. The presence of the cagA, vacA, and iceA genes were detected in 77.6%, 100%, and 80.4% of cases, respectively. Notably, cagA( +) with EPIYA-ABD, vacA s1i1m1, vacA s1i1m2, iceA1, and iceA2 accounted for 73.8%, 44.9%, 33.6%, 48.6%, and 31.8% of cases, respectively. Four iceA2 subtypes (24-aa, 59-aa, 94-aa, and 129-aa variants) were found, with the 59-aa variant the most prevalent (70.6%). The cagA( +)/vacAs1i1m1/iceA1 and cagA( +)/vacAs1i1m2/iceA1 combinations were found in 26.2% and 25.1% of cases, respectively. A multivariable logistic regression analysis was performed, after adjusting for age and gender, with the gastritis group was used as a reference control. Statistically significant associations were found between the vacA s1i1m2 genotype, the iceA1 variant, and the cagA( +)/vacAs1i1m2/iceA1 combination and gastric cancer; the adjusted ORs were estimated as 18.02 (95% CI: 3.39-95.81), 4.09 (95% CI: 1.1-15.08), and 16.19 (95% CI: 3.42-76.66), respectively. Interestingly, for the first time, our study found that vacA s1i1m2, but not vacA s1i1m1, was a risk factor for gastric cancer. This study illustrates the genetic diversity of the H. pylori cagA, vacA, and iceA genes across geographical regions and contributes to understanding the importance of these genotypes for clinical outcomes.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Genotype , Helicobacter Infections , Helicobacter pylori , Humans , Bacterial Proteins/genetics , Helicobacter pylori/genetics , Helicobacter pylori/isolation & purification , Helicobacter pylori/classification , Helicobacter pylori/pathogenicity , Vietnam/epidemiology , Antigens, Bacterial/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/epidemiology , Cross-Sectional Studies , Male , Female , Middle Aged , Adult , Bacterial Outer Membrane Proteins/genetics , Aged , Young Adult , Prevalence , Virulence Factors/genetics
18.
mSphere ; 9(5): e0000524, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38661343

ABSTRACT

The mycobacterial cell envelope is a major virulence determinant in pathogenic mycobacteria. Specific outer lipids play roles in pathogenesis, modulating the immune system and promoting the secretion of virulence factors. ESX-1 (ESAT-6 system-1) is a conserved protein secretion system required for mycobacterial pathogenesis. Previous studies revealed that mycobacterial strains lacking the outer lipid PDIM have impaired ESX-1 function during laboratory growth and infection. The mechanisms underlying changes in ESX-1 function are unknown. We used a proteo-genetic approach to measure phthiocerol dimycocerosate (PDIM)- and phenolic glycolipid (PGL)-dependent protein secretion in M. marinum, a non-tubercular mycobacterial pathogen that causes tuberculosis-like disease in ectothermic animals. Importantly, M. marinum is a well-established model for mycobacterial pathogenesis. Our findings showed that M. marinum strains without PDIM and PGL showed specific, significant reductions in protein secretion compared to the WT and complemented strains. We recently established a hierarchy for the secretion of ESX-1 substrates in four (I-IV) groups. Loss of PDIM differentially impacted secretion of Group III and IV ESX-1 substrates, which are likely the effectors of pathogenesis. Our data suggest that the altered secretion of specific ESX-1 substrates is responsible for the observed ESX-1-related effects in PDIM-deficient strains.IMPORTANCEMycobacterium tuberculosis, the cause of human tuberculosis, killed an estimated 1.3 million people in 2022. Non-tubercular mycobacterial species cause acute and chronic human infections. Understanding how these bacteria cause disease is critical. Lipids in the cell envelope are essential for mycobacteria to interact with the host and promote disease. Strains lacking outer lipids are attenuated for infection, but the reasons are unclear. Our research aims to identify a mechanism for attenuation of mycobacterial strains without the PDIM and PGL outer lipids in M. marinum. These findings will enhance our understanding of the importance of lipids in pathogenesis and how these lipids contribute to other established virulence mechanisms.


Subject(s)
Bacterial Proteins , Glycolipids , Mycobacterium marinum , Virulence Factors , Mycobacterium marinum/pathogenicity , Mycobacterium marinum/genetics , Mycobacterium marinum/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Glycolipids/metabolism , Virulence , Lipids , Antigens, Bacterial/metabolism , Antigens, Bacterial/genetics
19.
Microb Biotechnol ; 17(3): e14446, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38536702

ABSTRACT

Developing protein-based vaccines against bacteria has proved much more challenging than producing similar immunisations against viruses. Currently, anti-bacterial vaccines are designed using methods based on reverse vaccinology. These identify broadly conserved, immunogenic proteins using a combination of genomic and high-throughput laboratory data. While this approach has successfully generated multiple rationally designed formulations that show promising immunogenicity in animal models, few have been licensed. The difficulty of inducing protective immunity in humans with such vaccines mirrors the ability of many bacteria to recolonise individuals despite recognition by natural polyvalent antibody repertoires. As bacteria express too many antigens to evade all adaptive immune responses through mutation, they must instead inhibit the efficacy of such host defences through expressing surface structures that interface with the immune system. Therefore, 'immune interface interference' (I3) vaccines that target these features should synergistically directly target bacteria and prevent them from inhibiting responses to other surface antigens. This approach may help us understand the efficacy of the two recently introduced immunisations against serotype B meningococci, which both target the Factor H-binding protein (fHbp) that inhibits complement deposition on the bacterial surface. Therefore, I3 vaccine designs may help overcome the current challenges of developing protein-based vaccines to prevent bacterial infections.


Subject(s)
Meningococcal Vaccines , Neisseria meningitidis , Animals , Humans , Bacterial Vaccines/genetics , Bacterial Proteins/genetics , Antigens, Bacterial/genetics , Antibodies, Bacterial , Neisseria meningitidis/genetics
20.
Genes (Basel) ; 15(3)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38540398

ABSTRACT

Helicobacter pylori (H. pylori) is associated with gastric inflammation and mucosal antibodies against its cytotoxin-associated gene A (CagA) are protective. Vaccine-elicited immunity against H. pylori requires MHC class II expression, indicating that CD4+ T cells are protective. We hypothesized that the HLA-DR genotypes in human populations include protective alleles that more effectively bind immunogenic CagA peptide fragments and susceptible alleles with an impaired capacity to present CagA peptides. We recruited patients (n = 170) admitted for gastroendoscopy procedures and performed high-resolution HLA-DRB1 typing. Serum anti-CagA IgA levels were analyzed by ELISA (23.2% positive) and H. pylori classified as positive or negative in gastric mucosal tissue slides (72.9% positive). Pearson Chi-square analysis revealed that H. pylori infection was significantly increased in DRB1*11:04-positive individuals (p = 0.027). Anti-CagA IgA was significantly decreased in DRB1*11:04 positive individuals (p = 0.041). In contrast, anti-CagA IgA was significantly increased in DRB1*03:01 positive individuals (p = 0.030). For these HLA-DRB1 alleles of interest, we utilized two in silico prediction methods to compare their capacity to present CagA peptides. Both methods predicted increased numbers of peptides for DRB1*03:01 than DRB1*11:04. In addition, both alleles preferred distinctively different CagA 15mer peptide sequences for high affinity binding. These observations suggest that DRB1*11:04 is a susceptible genotype with impaired CagA immunity, whereas DRB1*03:01 is a protective genotype that promotes enhanced CagA immunity.


Subject(s)
Gastritis , Helicobacter pylori , Humans , Helicobacter pylori/genetics , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , HLA-DRB1 Chains/genetics , Cytotoxins , Gastritis/genetics , Genotype , Peptides/genetics , Immunoglobulin A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...