Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 640
Filter
1.
Front Biosci (Landmark Ed) ; 29(7): 246, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39082330

ABSTRACT

BACKGROUND: Pneumocystis jirovecii is the most emerging life-threating health problem that causes acute and fatal pneumonia infection. It is rare and more contagious for patients with leukemia and immune-deficiency disorders. Until now there is no treatment available for this infection therefore, it is needed to develop any treatment against this pathogen. METHODS: In this work, we used comparative proteomics, robust immune-informatics, and reverse vaccinology to create an mRNA vaccine against Pneumocystis jirovecii by targeting outer and transmembrane proteins. Using a comparative subtractive proteomic analysis of two Pneumocystis jirovecii proteomes, a distinct non-redundant Pneumocystis jirovecii (strain SE8) proteome was chosen. Seven Pneumocystis jirovecii transmembrane proteins were chosen from this proteome based on hydrophilicity, essentiality, virulence, antigenicity, pathway interaction, protein-protein network analysis, and allergenicity. OBJECTIVE: The reverse vaccinology approach was used to predict the immunogenic and antigenic epitopes of major histocompatibility complex (MHC) I, II and B-cells from the selected proteins on the basis of their antigenicity, toxicity and allergenicity. These immunogenic epitopes were linked together to construct the mRNA-based vaccine. To enhance the immunogenicity, suitable adjuvant, linkers (GPGPG, KK, and CYY), and PRDRE sequences were used. RESULTS: Through predictive modeling and confirmation via the Ramachandran plot, we assessed secondary and 3D structures. The adjuvant RpfE was incorporated to enhance the vaccine construct's immunogenicity (GRAVY index: -0.271, instability index: 39.53, antigenicity: 1.0428). The physiochemical profiling of vaccine construct was predicted it an antigenic, efficient, and potential vaccine. Notably, strong interactions were observed between the vaccine construct and TLR-3/TLR-4 (-1301.7 kcal/mol-1 and -1374.7 kcal/mol-1). CONCLUSIONS: The results predicted that mRNA-based vaccines trigger a cellular and humoral immune response, making the vaccine potential candidate against Pneumocystis jirovecii and it is more suitable for in-vitro analysis and validation to prove its effectiveness.


Subject(s)
Pneumocystis carinii , Pneumonia, Pneumocystis , Proteomics , Vaccinology , mRNA Vaccines , Proteomics/methods , Pneumocystis carinii/immunology , Pneumocystis carinii/genetics , Humans , Vaccinology/methods , mRNA Vaccines/immunology , Pneumonia, Pneumocystis/prevention & control , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/microbiology , Fungal Vaccines/immunology , Fungal Proteins/immunology , Fungal Proteins/genetics , Proteome/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccine Development/methods , Vaccines, Synthetic/immunology
2.
Sci Immunol ; 9(96): eadq7284, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38848339

ABSTRACT

Whole-proteome autoantibody profiling reveals an immunological signature that predates the clinical onset of multiple sclerosis.


Subject(s)
Autoantibodies , Biomarkers , Multiple Sclerosis , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/genetics , Autoantibodies/immunology , Autoantibodies/blood , Proteome/immunology , Proteomics/methods
3.
Front Immunol ; 15: 1413893, 2024.
Article in English | MEDLINE | ID: mdl-38915396

ABSTRACT

Introduction: Trypanosoma cruzi is a protozoan parasite that causes the tropical ailment known as Chagas disease, which has its origins in South America. Globally, it has a major impact on health and is transported by insect vector that serves as a parasite. Given the scarcity of vaccines and the limited treatment choices, we conducted a comprehensive investigation of core proteomics to explore a potential reverse vaccine candidate with high antigenicity. Methods: To identify the immunodominant epitopes, T. cruzi core proteomics was initially explored. Consequently, the vaccine sequence was engineered to possess characteristics of non-allergenicity, antigenicity, immunogenicity, and enhanced solubility. After modeling the tertiary structure of the human TLR4 receptor, the binding affinities were assessed employing molecular docking and molecular dynamics simulations (MDS). Results: Docking of the final vaccine design with TLR4 receptors revealed substantial hydrogen bond interactions. A server-based methodology for immunological simulation was developed to forecast the effectiveness against antibodies (IgM + IgG) and interferons (IFN-g). The MDS analysis revealed notable levels of structural compactness and binding stability with average RMSD of 5.03 Aring;, beta-factor 1.09e+5 Å, Rg is 44.7 Aring; and RMSF of 49.50 Aring;. This is followed by binding free energies calculation. The system stability was compromised by the complexes, as evidenced by their corresponding Gibbs free energies of -54.6 kcal/mol. Discussion: Subtractive proteomics approach was applied to determine the antigenic regions of the T cruzi. Our study utilized computational techniques to identify B- and T-cell epitopes in the T. cruzi core proteome. In current study the developed vaccine candidate exhibits immunodominant features. Our findings suggest that formulating a vaccine targeting the causative agent of Chagas disease should be the initial step in its development.


Subject(s)
Chagas Disease , Molecular Docking Simulation , Molecular Dynamics Simulation , Proteome , Protozoan Vaccines , Toll-Like Receptor 4 , Trypanosoma cruzi , Trypanosoma cruzi/immunology , Chagas Disease/immunology , Chagas Disease/prevention & control , Humans , Proteome/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Protozoan Vaccines/immunology , Animals , Immunodominant Epitopes/immunology , Proteomics/methods , Antigens, Protozoan/immunology , Antigens, Protozoan/chemistry , Antibodies, Protozoan/immunology , Protozoan Proteins/immunology , Protozoan Proteins/chemistry , Vaccine Development , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry
4.
Fish Shellfish Immunol ; 151: 109688, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38857817

ABSTRACT

This study marks the first utilization of reverse vaccinology to develop recombinant subunit vaccines against Pseudomonas koreensis infection in Empurau (Tor tambroides). The proteome (5538 proteins) was screened against various filters to prioritize proteins based on features that are associated with virulence, subcellular localization, transmembrane helical structure, antigenicity, essentiality, non-homology with the host proteome, molecular weight, and stability, which led to the identification of eight potential vaccine candidates. These potential vaccine candidates were cloned and expressed, with six achieving successful expression and purification. The antigens were formulated into two distinct vaccine mixtures, Vac A and Vac B, and their protective efficacy was assessed through in vivo challenge experiments. Vac A and Vac B demonstrated high protective efficacies of 100 % and 81.2 %, respectively. Histological analyses revealed reduced tissue damage in vaccinated fish after experimental infection, with Vac A showing no adverse effects, whereas Vac B exhibited mild degenerative changes. Quantitative real-time PCR results showed a significant upregulation of TNF-α and downregulation of IL-1ß in the kidneys, spleen, gills, and intestine in both Vac A- and Vac B-immunized fish after challenged with P. koreensis. Additionally, IL-8 exhibits tissue-specific differential expression, with significant upregulation in the kidney, gills, and intestine, and downregulation in the spleen, particularly notable in Vac A-immunized fish. The research underscores the effectiveness of the reverse vaccinology approach in fish and demonstrates the promising potential of Vac A and Vac B as recombinant subunit vaccines.


Subject(s)
Fish Diseases , Pseudomonas Infections , Pseudomonas , Animals , Fish Diseases/immunology , Fish Diseases/prevention & control , Pseudomonas/immunology , Pseudomonas Infections/veterinary , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccinology , Vaccines, Synthetic/immunology , Cyprinidae/immunology , Pseudomonas Vaccines/immunology , Proteome/immunology
5.
Front Immunol ; 15: 1395870, 2024.
Article in English | MEDLINE | ID: mdl-38799422

ABSTRACT

Emerging infectious diseases represent a significant threat to global health, with West Nile virus (WNV) being a prominent example due to its potential to cause severe neurological disorders alongside mild feverish conditions. Particularly prevalent in the continental United States, WNV has emerged as a global concern, with outbreaks indicating the urgent need for effective prophylactic measures. The current problem is that the absence of a commercial vaccine against WNV highlights a critical gap in preventive strategies against WNV. This study aims to address this gap by proposing a novel, multivalent vaccine designed using immunoinformatics approaches to elicit comprehensive humoral and cellular immune responses against WNV. The objective of the study is to provide a theoretical framework for experimental scientists to formulate of vaccine against WNV and tackle the current problem by generating an immune response inside the host. The research employs reverse vaccinology and subtractive proteomics methodologies to identify NP_041724.2 polyprotein and YP_009164950.1 truncated flavivirus polyprotein NS1 as the prime antigens. The selection process for epitopes focused on B and T-cell reactivity, antigenicity, water solubility, and non-allergenic properties, prioritizing candidates with the potential for broad immunogenicity and safety. The designed vaccine construct integrates these epitopes, connected via GPGPG linkers, and supplemented with an adjuvant with the help of another linker EAAAK, to enhance immunogenicity. Preliminary computational analyses suggest that the proposed vaccine could achieve near-universal coverage, effectively targeting approximately 99.74% of the global population, with perfect coverage in specific regions such as Sweden and Finland. Molecular docking and immune simulation studies further validate the potential efficacy of the vaccine, indicating strong binding affinity with toll-like receptor 3 (TLR-3) and promising immune response profiles, including significant antibody-mediated and cellular responses. These findings present the vaccine construct as a viable candidate for further development and testing. While the theoretical and computational results are promising, advancing from in-silico predictions to a tangible vaccine requires comprehensive laboratory validation. This next step is essential to confirm the vaccine's efficacy and safety in eliciting an immune response against WNV. Through this study, we propose a novel approach to vaccine development against WNV and contribute to the broader field of immunoinformatics, showcasing the potential to accelerate the design of effective vaccines against emerging viral threats. The journey from hypothesis to practical solution embodies the interdisciplinary collaboration essential for modern infectious disease management and prevention strategies.


Subject(s)
Computational Biology , Immunodominant Epitopes , Proteome , Vaccines, Subunit , West Nile Fever , West Nile Virus Vaccines , West Nile virus , West Nile virus/immunology , Immunodominant Epitopes/immunology , Humans , Proteome/immunology , West Nile Fever/prevention & control , West Nile Fever/immunology , West Nile Fever/virology , Computational Biology/methods , West Nile Virus Vaccines/immunology , Vaccines, Subunit/immunology , Vaccine Development , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Proteomics/methods , Immunoinformatics , Protein Subunit Vaccines
6.
Food Chem ; 451: 139295, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38729042

ABSTRACT

Information regarding protein expression and phosphorylation modifications in the bovine milk fat globule membrane is scarce, particularly throughout various lactation periods. This study employed a complete proteome and phosphoproteome between bovine colostrum and mature milk. A total of 11 proteins were seen in both protein expression and phosphorylation levels. There were 400 proteins identified in only protein expression, and 104 phosphoproteins identified in only phosphorylation levels. A total of 232 significant protein characteristics were identified within the proteome and significant phosphorylation sites within 86 phosphoproteins of the phosphoproteome. Biological activities and pathways primarily exhibited associations with the immune system. Simultaneously, a comprehensive analysis of proteins and phosphorylation sites using a multi-omics approach. Hence, the data we have obtained has the potential to expand our understanding of how the bovine milk fat globule membrane might be utilized as a beneficial component in dairy products.


Subject(s)
Glycolipids , Glycoproteins , Lactation , Lipid Droplets , Milk , Phosphoproteins , Proteomics , Animals , Cattle , Glycoproteins/chemistry , Glycoproteins/immunology , Glycoproteins/metabolism , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Glycolipids/chemistry , Glycolipids/metabolism , Female , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/immunology , Milk/chemistry , Milk Proteins/chemistry , Milk Proteins/metabolism , Milk Proteins/immunology , Phosphorylation , Proteome/chemistry , Proteome/immunology , Proteome/analysis
7.
Nature ; 628(8006): 171-179, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509360

ABSTRACT

The myriad microorganisms that live in close association with humans have diverse effects on physiology, yet the molecular bases for these impacts remain mostly unknown1-3. Classical pathogens often invade host tissues and modulate immune responses through interactions with human extracellular and secreted proteins (the 'exoproteome'). Commensal microorganisms may also facilitate niche colonization and shape host biology by engaging host exoproteins; however, direct exoproteome-microbiota interactions remain largely unexplored. Here we developed and validated a novel technology, BASEHIT, that enables proteome-scale assessment of human exoproteome-microbiome interactions. Using BASEHIT, we interrogated more than 1.7 million potential interactions between 519 human-associated bacterial strains from diverse phylogenies and tissues of origin and 3,324 human exoproteins. The resulting interactome revealed an extensive network of transkingdom connectivity consisting of thousands of previously undescribed host-microorganism interactions involving 383 strains and 651 host proteins. Specific binding patterns within this network implied underlying biological logic; for example, conspecific strains exhibited shared exoprotein-binding patterns, and individual tissue isolates uniquely bound tissue-specific exoproteins. Furthermore, we observed dozens of unique and often strain-specific interactions with potential roles in niche colonization, tissue remodelling and immunomodulation, and found that strains with differing host interaction profiles had divergent interactions with host cells in vitro and effects on the host immune system in vivo. Overall, these studies expose a previously unexplored landscape of molecular-level host-microbiota interactions that may underlie causal effects of indigenous microorganisms on human health and disease.


Subject(s)
Bacteria , Host Microbial Interactions , Microbiota , Phylogeny , Proteome , Symbiosis , Animals , Female , Humans , Mice , Bacteria/classification , Bacteria/immunology , Bacteria/metabolism , Bacteria/pathogenicity , Host Microbial Interactions/immunology , Host Microbial Interactions/physiology , Host Tropism , Microbiota/immunology , Microbiota/physiology , Organ Specificity , Protein Binding , Proteome/immunology , Proteome/metabolism , Reproducibility of Results
8.
Biomed Res Int ; 2023: 6325568, 2023.
Article in English | MEDLINE | ID: mdl-37415928

ABSTRACT

Antibiotic resistance is a major public health concern that has resulted in high healthcare costs, increased mortality, and the emergence of novel bacterial diseases. Cardiobacterium valvarum, an antibiotic-resistant bacterium, is one of the leading causes of heart disease. Currently, there is no licensed vaccination against C. valvarum. In this research, an in silico-based vaccine was designed against C. valvarum using reverse vaccinology, bioinformatics, and immunoinformatics techniques. 4206 core proteins, 2027 nonredundant proteins, and 2179 redundant proteins were predicted. Among nonredundant proteins, 23 proteins were predicted in an extracellular membrane, 30 in the outer membrane, and 62 in the periplasmic membrane region. After applying several subtractive proteomics filters, two proteins, TonB-dependent siderophore receptor and hypothetical protein, were chosen for epitope prediction. In the epitope selection phase, B and T-cellepitopes were analyzed and shortlisted for vaccine design. The vaccine model was designed by linking selected epitopes with GPGPG linkers to avoid flexibility. Furthermore, the vaccine model was linked to cholera toxin B adjuvant to induce a proper immune response. The docking approach was utilized to analyze binding affinity to immune cell receptors. Molecular docking results predicted 12.75 kcal/mol for a Vaccine with MHC-I, 6.89 for a vaccine with MHC-II, and 19.51 vaccine with TLR-4. The MMGBSA estimated -94, -78, and -76 kcal/mol for TLR-4 and vaccine, MHC-I and vaccine, and MHC-II and vaccine, while the MMPBSA analysis estimated -97, -61, and -72 kcal/mol for TLR-4 with the vaccine, MHC-I with vaccine, and MHC-II with a vaccine. Molecular dynamic simulation analysis revealed that the designed vaccine construct has proper stability with immune cell receptors as it is essential for inducing an immune response. In conclusion, we observed that the model vaccine candidate has the potency to induce an immune response in the host. However, the study is designed purely on a computational basis; hence, experimental validation is strongly recommended.


Subject(s)
Bacterial Vaccines , Molecular Docking Simulation , Proteome/immunology , Bacterial Proteins/immunology , Epitopes/immunology , T-Lymphocytes/immunology
9.
Vet Res ; 54(1): 32, 2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37016420

ABSTRACT

Host response to invasive microbes in the bovine udder has an important role on the animal health and is essential to the dairy industry to ensure production of high-quality milk and reduce the mastitis incidence. To better understand the biology behind these host-microbiome interactions, we investigated the somatic cell proteomes at quarter level for four cows (collected before and after milking) using a shotgun proteomics approach. Simultaneously, we identified the quarter microbiota by amplicon sequencing to detect presence of mastitis pathogens or other commensal taxa. In total, 32 quarter milk samples were analyzed divided in two groups depending on the somatic cell count (SCC). The high SCC group (>100,000 cell/mL) included 10 samples and significant different proteome profiles were detected. Differential abundance analysis uncovers a specific expression pattern in high SCC samples revealing pathways involved in immune responses such as inflammation, activation of the complement system, migration of immune cells, and tight junctions. Interestingly, different proteome profiles were also identified in quarter samples containing one of the two mastitis pathogens, Staphylococcus aureus and Streptococcus uberis, indicating a different response of the host depending on the pathogen. Weighted correlation network analysis identified three modules of co-expressed proteins which were correlated with the SCC in the quarters. These modules contained proteins assigned to different aspects of the immune response, but also amino sugar and nucleotide sugar metabolism, and biosynthesis of amino acids. The results of this study provide deeper insights on how the proteome expression changes at quarter level in naturally infected cows and pinpoint potential interactions and important biological functions during host-microbe interaction.


Subject(s)
Host Microbial Interactions , Mammary Glands, Animal , Milk , Proteome , Animals , Cattle , Female , Cattle Diseases/immunology , Cattle Diseases/microbiology , Cell Count/veterinary , Mammary Glands, Animal/immunology , Mammary Glands, Animal/microbiology , Mastitis, Bovine/immunology , Mastitis, Bovine/microbiology , Milk/cytology , Proteome/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/veterinary , Host Microbial Interactions/immunology
10.
Science ; 379(6637): 1092-1093, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36927028
11.
Immunol Res ; 71(2): 247-266, 2023 04.
Article in English | MEDLINE | ID: mdl-36459272

ABSTRACT

Brucella suis mediates the transmission of brucellosis in humans and animals and a significant facultative zoonotic pathogen found in livestock. It has the capacity to survive and multiply in a phagocytic environment and to acquire resistance under hostile conditions thus becoming a threat globally. Antibiotic resistance is posing a substantial public health threat, hence there is an unmet and urgent clinical need for immune-based non-antibiotic methods to treat brucellosis. Hence, we aimed to explore the whole proteome of Brucella suis to predict antigenic proteins as a vaccine target and designed a novel chimeric vaccine (multi-epitope vaccine) through subtractive genomics-based reverse vaccinology approaches. The applied subsequent hierarchical shortlisting resulted in the identification of Multidrug efflux Resistance-nodulation-division (RND) transporter outer membrane subunit (gene BepC) that may act as a potential vaccine target. T-cell and B-cell epitopes have been predicted from target proteins using a number of immunoinformatic methods. Six MHC I, ten MHC II, and four B-cell epitopes were used to create a 324-amino-acid MEV construct, which was coupled with appropriate linkers and adjuvant. To boost the immunological response to the vaccine, the vaccine was combined with the TLR4 agonist HBHA protein. The MEV structure predicted was found to be highly antigenic, non-toxic, non-allergenic, flexible, stable, and soluble. To confirm the interactions with the receptors, a molecular docking simulation of the MEV was done using the human TLR4 (toll-like receptor 4) and HLAs. The stability and binding of the MEV-docked complexes with TLR4 were assessed using molecular dynamics (MD) simulation. Finally, MEV was reverse translated, its cDNA structure was evaluated, and then, in silico cloning into an E. coli expression host was conducted to promote maximum vaccine protein production with appropriate post-translational modifications. These comprehensive computer calculations backed up the efficacy of the suggested MEV in protecting against B. suis infections. However, more experimental validations are needed to adequately assess the vaccine candidate's potential. HIGHLIGHTS: • Subtractive genomic analysis and reverse vaccinology for the prioritization of novel vaccine target • Examination of chimeric vaccine in terms of allergenicity, antigenicity, MHC I, II binding efficacy, and structural-based studies • Molecular docking simulation method to rank based vaccine candidate and understand their binding modes.


Subject(s)
Brucella Vaccine , Brucella suis , Brucellosis , Animals , Humans , Brucella suis/genetics , Brucella suis/immunology , Brucellosis/genetics , Brucellosis/immunology , Brucellosis/prevention & control , Computational Biology , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte , Escherichia coli , Molecular Docking Simulation , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/immunology , Proteome/genetics , Proteome/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Brucella Vaccine/genetics , Brucella Vaccine/immunology , Brucella Vaccine/therapeutic use , Epitopes/genetics , Epitopes/immunology , Vaccine Development , Drug Design
12.
Nature ; 608(7922): 397-404, 2022 08.
Article in English | MEDLINE | ID: mdl-35922511

ABSTRACT

The human immune system is composed of a distributed network of cells circulating throughout the body, which must dynamically form physical associations and communicate using interactions between their cell-surface proteomes1. Despite their therapeutic potential2, our map of these surface interactions remains incomplete3,4. Here, using a high-throughput surface receptor screening method, we systematically mapped the direct protein interactions across a recombinant library that encompasses most of the surface proteins that are detectable on human leukocytes. We independently validated and determined the biophysical parameters of each novel interaction, resulting in a high-confidence and quantitative view of the receptor wiring that connects human immune cells. By integrating our interactome with expression data, we identified trends in the dynamics of immune interactions and constructed a reductionist mathematical model that predicts cellular connectivity from basic principles. We also developed an interactive multi-tissue single-cell atlas that infers immune interactions throughout the body, revealing potential functional contexts for new interactions and hubs in multicellular networks. Finally, we combined targeted protein stimulation of human leukocytes with multiplex high-content microscopy to link our receptor interactions to functional roles, in terms of both modulating immune responses and maintaining normal patterns of intercellular associations. Together, our work provides a systematic perspective on the intercellular wiring of the human immune system that extends from systems-level principles of immune cell connectivity down to mechanistic characterization of individual receptors, which could offer opportunities for therapeutic intervention.


Subject(s)
Cell Communication , Immune System , Protein Interaction Maps , Cell Communication/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Leukocytes/chemistry , Leukocytes/immunology , Leukocytes/metabolism , Protein Binding , Proteome/immunology , Proteome/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism
13.
Proc Natl Acad Sci U S A ; 119(13): e2117770119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35312359

ABSTRACT

Spirochetal pathogens, such as the causative agent of Lyme disease, Borrelia burgdorferi sensu lato, encode an abundance of lipoproteins; however, due in part to their evolutionary distance from more well-studied bacteria, such as Proteobacteria and Firmicutes, few spirochetal lipoproteins have assigned functions. Indeed, B. burgdorferi devotes almost 8% of its genome to lipoprotein genes and interacts with its environment primarily through the production of at least 80 surface-exposed lipoproteins throughout its tick vector­vertebrate host lifecycle. Several B. burgdorferi lipoproteins have been shown to serve roles in cellular adherence or immune evasion, but the functions for most B. burgdorferi surface lipoproteins remain unknown. In this study, we developed a B. burgdorferi lipoproteome screening platform utilizing intact spirochetes that enables the identification of previously unrecognized host interactions. As spirochetal survival in the bloodstream is essential for dissemination, we targeted our screen to C1, the first component of the classical (antibody-initiated) complement pathway. We identified two high-affinity C1 interactions by the paralogous lipoproteins, ElpB and ElpQ (also termed ErpB and ErpQ, respectively). Using biochemical, microbiological, and biophysical approaches, we demonstrate that ElpB and ElpQ bind the activated forms of the C1 proteases, C1r and C1s, and represent a distinct mechanistic class of C1 inhibitors that protect the spirochete from antibody-mediated complement killing. In addition to identifying a mode of complement inhibition, our study establishes a lipoproteome screening methodology as a discovery platform for identifying direct host­pathogen interactions that are central to the pathogenesis of spirochetes, such as the Lyme disease agent.


Subject(s)
Bacterial Proteins , Borrelia burgdorferi , Complement C1q , Immune Evasion , Lipoproteins , Lyme Disease , Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement C1q/immunology , Humans , Immunoglobulins/immunology , Lipoproteins/immunology , Lyme Disease/immunology , Lyme Disease/microbiology , Proteome/immunology
14.
J Gen Virol ; 103(1)2022 01.
Article in English | MEDLINE | ID: mdl-35014605

ABSTRACT

The pandemic caused by SARS-CoV-2 has led to the successful development of effective vaccines however the prospect of variants of SARS-CoV-2 and future coronavirus outbreaks necessitates the investigation of other vaccine strategies capable of broadening vaccine mediated T-cell responses and potentially providing cross-immunity. In this study the SARS-CoV-2 proteome was assessed for clusters of immunogenic epitopes restricted to diverse human leucocyte antigen. These regions were then assessed for their conservation amongst other coronaviruses representative of different alpha and beta coronavirus genera. Sixteen highly conserved peptides containing numerous HLA class I and II restricted epitopes were synthesized from these regions and assessed in vitro for their antigenicity against T-cells from individuals with previous SARS-CoV-2 infection. Monocyte derived dendritic cells were generated from these peripheral blood mononuclear cells (PBMC), loaded with SARS-CoV-2 peptides, and used to induce autologous CD4+ and CD8+ T cell activation. The SARS-CoV-2 peptides demonstrated antigenicity against the T-cells from individuals with previous SARS-CoV-2 infection indicating that this approach holds promise as a method to activate anti-SAR-CoV-2 T-cell responses from conserved regions of the virus which are not included in vaccines utilising the Spike protein.


Subject(s)
Peptides/immunology , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , COVID-19 Vaccines , Coronavirus/classification , Coronavirus/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Peptides/chemical synthesis , Peptides/chemistry , Proteome/immunology , Vaccines, Subunit , Viral Proteins/immunology
15.
Cell Rep ; 38(3): 110271, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35026155

ABSTRACT

The utility of the urinary proteome in infectious diseases remains unclear. Here, we analyzed the proteome and metabolome of urine and serum samples from patients with COVID-19 and healthy controls. Our data show that urinary proteins effectively classify COVID-19 by severity. We detect 197 cytokines and their receptors in urine, but only 124 in serum using TMT-based proteomics. The decrease in urinary ESCRT complex proteins correlates with active SARS-CoV-2 replication. The downregulation of urinary CXCL14 in severe COVID-19 cases positively correlates with blood lymphocyte counts. Integrative multiomics analysis suggests that innate immune activation and inflammation triggered renal injuries in patients with COVID-19. COVID-19-associated modulation of the urinary proteome offers unique insights into the pathogenesis of this disease. This study demonstrates the added value of including the urinary proteome in a suite of multiomics analytes in evaluating the immune pathobiology and clinical course of COVID-19 and, potentially, other infectious diseases.


Subject(s)
COVID-19/urine , Immunity , Metabolome , Proteome/analysis , SARS-CoV-2/immunology , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/blood , COVID-19/immunology , COVID-19/pathology , Case-Control Studies , Child , Child, Preschool , China , Cohort Studies , Female , Humans , Immunity/physiology , Male , Metabolome/immunology , Metabolomics , Middle Aged , Patient Acuity , Proteome/immunology , Proteome/metabolism , Proteomics , Urinalysis/methods , Young Adult
16.
J Clin Lab Anal ; 36(1): e24133, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34812532

ABSTRACT

OBJECTIVE: The diagnosis of seronegative rheumatoid arthritis (SNRA) is often difficult due to the unavailability of reliable laboratory markers. The aim of this study was to identify differentially expressed proteins in sera of SNRA, seropositive RA (SPRA), and healthy donors (HD). METHODS: A total of 32 seropositive RA patients, 32 SNRA patients, and 35 HD were enrolled in our study. Differentially expressed proteins between 3 groups were identified via isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomic analysis, and an ELISA test was used for the validation test. Correlation analysis was conducted by GraphPad Prism. RESULTS: Using iTRAQ quantitative proteomics, we identified 14 proteins were significantly different between SPRA and SNRA, including 4 upregulated proteins and 10 downregulated proteins. Four differentially expressed proteins were validated by ELISA test, and the results showed that SAA1 protein was significantly higher in SPRA and SNRA patients compared with HD, and PSME1 was elevated in SPRA patients. What's more, SAA1 was increased in the anti-CCP or RF high-level group in RA patients, and PSME1 was increased in the RF high-level group. Alternatively, SAA1 was positively correlated with inflammation indicators in RA patients, while PSME1 showed no correlation with inflammation indicators. CONCLUSIONS: iTRAQ proteomic approaches revealed variations in serum protein composition among SPRA patients, SNRA patients, and HD and provided new idea for advanced diagnostic methods and precision treatment of RA.


Subject(s)
Arthritis, Rheumatoid , Proteome/analysis , Proteomics , Adult , Aged , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoantibodies/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Isotope Labeling , Male , Mass Spectrometry , Middle Aged , Proteome/chemistry , Proteome/immunology
17.
Front Immunol ; 12: 789748, 2021.
Article in English | MEDLINE | ID: mdl-34938298

ABSTRACT

Recently, a mass spectrometry-based approach was introduced to directly assess the IgG1 immunoglobulin clonal repertoires in plasma. Here we expanded upon this approach by describing a mass spectrometry-based technique to assess specifically the clonal repertoire of another important class of immunoglobulin molecules, IgA1, and show it is efficiently and robustly applicable to either milk or plasma samples. Focusing on two individual healthy donors, whose milk was sampled longitudinally during the first 16 weeks of lactation, we demonstrate that the total repertoire of milk sIgA1 is dominated by only 50-500 clones, even though the human body theoretically can generate several orders of magnitude more clones. We show that in each donor the sIgA1 repertoire only changes marginally and quite gradually over the monitored 16-week period of lactation. Furthermore, the observed overlap in clonal repertoires between the two individual donors is close to non-existent. Mothers provide protection to their newborn infants directly by the transfer of antibodies via breastfeeding. The approach introduced here, can be used to visualize the clonal repertoire transferred from mother to infant and to detect changes in-time in that repertoire adapting to changes in maternal physiology.


Subject(s)
Immunoglobulin A, Secretory/immunology , Mass Spectrometry , Milk, Human/immunology , Proteome/immunology , Proteomics , Breast Milk Expression , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Colostrum/immunology , Colostrum/metabolism , Female , Humans , Immunoglobulin A, Secretory/blood , Lactation , Milk, Human/metabolism
18.
Front Immunol ; 12: 765898, 2021.
Article in English | MEDLINE | ID: mdl-34858420

ABSTRACT

Immunotherapies are revolutionizing cancer care, producing durable responses and potentially cures in a subset of patients. However, response rates are low for most tumors, grade 3/4 toxicities are not uncommon, and our current understanding of tumor immunobiology is incomplete. While hundreds of immunomodulatory proteins in the tumor microenvironment shape the anti-tumor response, few of them can be reliably quantified. To address this need, we developed a multiplex panel of targeted proteomic assays targeting 52 peptides representing 46 proteins using peptide immunoaffinity enrichment coupled to multiple reaction monitoring-mass spectrometry. We validated the assays in tissue and plasma matrices, where performance figures of merit showed over 3 orders of dynamic range and median inter-day CVs of 5.2% (tissue) and 21% (plasma). A feasibility study in clinical biospecimens showed detection of 48/52 peptides in frozen tissue and 38/52 peptides in plasma. The assays are publicly available as a resource for the research community.


Subject(s)
Chromatography, Liquid/methods , Mass Spectrometry/methods , Peptides/analysis , Proteome/analysis , Proteomics/methods , Specimen Handling/methods , Antibodies/analysis , Antibodies/immunology , Blotting, Western , Cell Line, Tumor , HeLa Cells , Humans , Jurkat Cells , MCF-7 Cells , Peptides/blood , Peptides/immunology , Proteome/genetics , Proteome/immunology , RNA-Seq/methods , Reproducibility of Results
19.
Microbiol Spectr ; 9(3): e0081421, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34935429

ABSTRACT

The molecular details underlying differences in pathogenicity between Rickettsia species remain to be fully understood. Evidence points to macrophage permissiveness as a key mechanism in rickettsial virulence. Different studies have shown that several rickettsial species responsible for mild forms of rickettsioses can also escape macrophage-mediated killing mechanisms and establish a replicative niche within these cells. However, their manipulative capacity with respect to host cellular processes is far from being understood. A deeper understanding of the interplay between mildly pathogenic rickettsiae and macrophages and the commonalities and specificities of host responses to infection would illuminate differences in immune evasion mechanisms and pathogenicity. We used quantitative proteomics by sequential windowed data independent acquisition of the total high-resolution mass spectra with tandem mass spectrometry (SWATH-MS/MS) to profile alterations resulting from infection of THP-1 macrophages with three mildly pathogenic rickettsiae: Rickettsia parkeri, Rickettsia africae, and Rickettsia massiliae, all successfully proliferating in these cells. We show that all three species trigger different proteome signatures. Our results reveal a significant impact of infection on proteins categorized as type I interferon responses, which here included several components of the retinoic acid-inducible gene I (RIG-1)-like signaling pathway, mRNA splicing, and protein translation. Moreover, significant differences in protein content between infection conditions provide evidence for species-specific induced alterations. Indeed, we confirm distinct impacts on host inflammatory responses between species during infection, demonstrating that these species trigger different levels of beta interferon (IFN-ß), differences in the bioavailability of the proinflammatory cytokine interleukin 1ß (IL-1ß), and differences in triggering of pyroptotic events. This work reveals novel aspects and exciting nuances of macrophage-Rickettsia interactions, adding additional layers of complexity between Rickettsia and host cells' constant arms race for survival. IMPORTANCE The incidence of diseases caused by Rickettsia has been increasing over the years. It has long been known that rickettsioses comprise diseases with a continuous spectrum of severity. There are highly pathogenic species causing diseases that are life threatening if untreated, others causing mild forms of the disease, and a third group for which no pathogenicity to humans has been described. These marked differences likely reflect distinct capacities for manipulation of host cell processes, with macrophage permissiveness emerging as a key virulence trait. However, what defines pathogenicity attributes among rickettsial species is far from being resolved. We demonstrate that the mildly pathogenic Rickettsia parkeri, Rickettsia africae, and Rickettsia massiliae, all successfully proliferating in macrophages, trigger different proteome signatures in these cells and differentially impact critical components of innate immune responses by inducing different levels of beta interferon (IFN-ß) and interleukin 1ß (IL-1ß) and different timing of pyroptotic events during infection. Our work reveals novel nuances in rickettsia-macrophage interactions, offering new clues to understand Rickettsia pathogenicity.


Subject(s)
Inflammation , Macrophages/microbiology , Proteins/genetics , Proteome/genetics , Rickettsia Infections/immunology , Rickettsia/immunology , Humans , Immune Evasion , Macrophages/immunology , Proteins/immunology , Proteome/immunology , Rickettsia/classification , Rickettsia/genetics , Rickettsia/physiology , Rickettsia Infections/genetics , Rickettsia Infections/microbiology
20.
Mol Cell Proteomics ; 20: 100155, 2021.
Article in English | MEDLINE | ID: mdl-34597790

ABSTRACT

Probing the human proteome in tissues and biofluids such as plasma is attractive for biomarker and drug target discovery. Recent breakthroughs in multiplex, antibody-based, proteomics technologies now enable the simultaneous quantification of thousands of proteins at as low as sub fg/ml concentrations with remarkable dynamic ranges of up to 10-log. We herein provide a comprehensive guide to the methodologies, performance, technical comparisons, advantages, and disadvantages of established and emerging technologies for the multiplexed ultrasensitive measurement of proteins. Gaining holistic knowledge on these innovations is crucial for choosing the right multiplexed proteomics tool for applications at hand to critically complement traditional proteomics methods. This can bring researchers closer than ever before to elucidating the intricate inner workings and cross talk that spans multitude of proteins in disease mechanisms.


Subject(s)
Antibodies/immunology , Proteome/analysis , Proteomics/methods , Humans , Immunoassay , Proteins/analysis , Proteins/immunology , Proteome/immunology
SELECTION OF CITATIONS
SEARCH DETAIL