Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.985
Filter
1.
Nat Commun ; 15(1): 6766, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117651

ABSTRACT

Live vaccines are ideal for inducing immunity but suffer from the need to attenuate their pathogenicity or replication to preclude the possibility of escape. Unnatural amino acids (UAAs) provide a strategy to engineer stringent auxotrophies, yielding conditionally replication incompetent live bacteria with excellent safety profiles. Here, we engineer Pseudomonas aeruginosa to maintain auxotrophy for the UAA p-benzoyl-L-phenylalanine (BzF) through its incorporation into the essential protein DnaN. In vivo evolution using an Escherichia coli-based two-hybrid selection system enabled engineering of a mutant DnaN homodimeric interface completely dependent on a BzF-specific interaction. This engineered strain, Pa Vaccine, exhibits undetectable escape frequency (<10-11) and shows excellent safety in naïve mice. Animals vaccinated via intranasal or intraperitoneal routes are protected from lethal challenge with pathogenic P. aeruginosa PA14. These results establish UAA-auxotrophic bacteria as promising candidates for bacterial vaccine therapy and outline a platform for expanding this technology to diverse bacterial pathogens.


Subject(s)
Pseudomonas Infections , Pseudomonas Vaccines , Pseudomonas aeruginosa , Animals , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Mice , Female , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/genetics , Amino Acids , Phenylalanine/analogs & derivatives , Escherichia coli/immunology , Escherichia coli/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Mice, Inbred BALB C
2.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39133098

ABSTRACT

Pseudomonas aeruginosa is a complex nosocomial infectious agent responsible for numerous illnesses, with its growing resistance variations complicating treatment development. Studies have emphasized the importance of virulence factors OprE and OprF in pathogenesis, highlighting their potential as vaccine candidates. In this study, B-cell, MHC-I, and MHC-II epitopes were identified, and molecular linkers were active to join these epitopes with an appropriate adjuvant to construct a vaccine. Computational tools were employed to forecast the tertiary framework, characteristics, and also to confirm the vaccine's composition. The potency was weighed through population coverage analysis and immune simulation. This project aims to create a multi-epitope vaccine to reduce P. aeruginosa-related illness and mortality using immunoinformatics resources. The ultimate complex has been determined to be stable, soluble, antigenic, and non-allergenic upon inspection of its physicochemical and immunological properties. Additionally, the protein exhibited acidic and hydrophilic characteristics. The Ramachandran plot, ProSA-web, ERRAT, and Verify3D were employed to ensure the final model's authenticity once the protein's three-dimensional structure had been established and refined. The vaccine model showed a significant binding score and stability when interacting with MHC receptors. Population coverage analysis indicated a global coverage rate of 83.40%, with the USA having the highest coverage rate, exceeding 90%. Moreover, the vaccine sequence underwent codon optimization before being cloned into the Escherichia coli plasmid vector pET-28a (+) at the EcoRI and EcoRV restriction sites. Our research has developed a vaccine against P. aeruginosa that has strong binding affinity and worldwide coverage, offering an acceptable way to mitigate nosocomial infections.


Subject(s)
Computational Biology , Pseudomonas Infections , Pseudomonas aeruginosa , Sepsis , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Humans , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Sepsis/prevention & control , Sepsis/immunology , Sepsis/microbiology , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Pneumonia/prevention & control , Pneumonia/immunology , Pneumonia/microbiology , Pseudomonas Vaccines/immunology , Bacterial Vaccines/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics
3.
Arch Dermatol Res ; 316(8): 548, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162738

ABSTRACT

The skin, being the body's largest organ, primarily functions as a formidable defense mechanism against potential microbial infections. The skin's microbiota, consisting of a complex assembly of microorganisms, exerts a pivotal influence on skin homeostasis by modulating keratinocytes and their cytokine secretion, thereby playing an integral role in promoting optimal cutaneous health. Leuconostoc mesenteroides finds extensive application in the production of fermented foods and bacteriocins. Empirical studies validate the effectiveness of L. mesenteroides treatments in enhancing immune function and demonstrating notable antioxidant characteristics. This study investigates the potential of L. mesenteroides in improving skin health and wound healing. It also aims to comprehend their impact on wound healing markers, cytokine production, and cell cycle regulation compared to ferulic acid, known for its wound healing effects. Our findings indicate that L. mesenteroides lysate possesses antibacterial properties against Staphylococcus aureus and Pseudomonas aeruginosa, along with the ability to mitigate their toxic effects in a pathogen-simulating model employing HaCaT keratinocyte cells. Additionally, the lysate demonstrated noteworthy wound closure after a 24-hour treatment, along with a significant reduction in interleukin-6 levels and oxidative stress index. Modulation of the cell cycle is evident by decreasing G0/G1 phases and increasing S and G2/M phases and enhanced expression of wound healing marker genes and proteins CDH1. In conclusion, L. mesenteroides lysate exhibits immune-modulating and antibacterial properties, offering potential alternatives to conventional treatments for various skin conditions. These findings contribute to the exploration of innovative approaches to enhancing human life through skin health and wound healing.


Subject(s)
HaCaT Cells , Keratinocytes , Leuconostoc mesenteroides , Pseudomonas aeruginosa , Staphylococcus aureus , Wound Healing , Keratinocytes/immunology , Humans , Wound Healing/drug effects , Wound Healing/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology , Leuconostoc mesenteroides/immunology , Leuconostoc mesenteroides/metabolism , Pseudomonas aeruginosa/immunology , Anti-Bacterial Agents/pharmacology , Skin/immunology , Skin/microbiology , Skin/pathology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Cell Cycle/drug effects , Antioxidants/pharmacology , Cell Line , Cytokines/metabolism , Interleukin-6/metabolism
5.
Cell Host Microbe ; 32(8): 1427-1443.e8, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39094584

ABSTRACT

Prokaryotes have evolved a multitude of defense systems to protect against phage predation. Some of these resemble eukaryotic genes involved in antiviral responses. Here, we set out to systematically project the current knowledge of eukaryotic-like antiviral defense systems onto prokaryotic genomes, using Pseudomonas aeruginosa as a model organism. Searching for phage defense systems related to innate antiviral genes from vertebrates and plants, we uncovered over 450 candidates. We validated six of these phage defense systems, including factors preventing viral attachment, R-loop-acting enzymes, the inflammasome, ubiquitin pathway, and pathogen recognition signaling. Collectively, these defense systems support the concept of deep evolutionary links and shared antiviral mechanisms between prokaryotes and eukaryotes.


Subject(s)
Pseudomonas aeruginosa , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/virology , Immunity, Innate , Bacteriophages/genetics , Bacteriophages/physiology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Animals , Evolution, Molecular , Inflammasomes/immunology , Inflammasomes/genetics , Eukaryota/virology , Eukaryota/genetics , Eukaryota/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Evolution , Plants/immunology , Plants/virology , Plants/microbiology
6.
Respir Res ; 25(1): 262, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951782

ABSTRACT

BACKGROUND: Donor-specific antibodies (DSAs) are common following lung transplantation (LuTx), yet their role in graft damage is inconclusive. Mean fluorescent intensity (MFI) is the main read-out of DSA diagnostics; however its value is often disregarded when analyzing unwanted post-transplant outcomes such as graft loss or chronic lung allograft dysfunction (CLAD). Here we aim to evaluate an MFI stratification method in these outcomes. METHODS: A cohort of 87 LuTx recipients has been analyzed, in which a cutoff of 8000 MFI has been determined for high MFI based on clinically relevant data. Accordingly, recipients were divided into DSA-negative, DSA-low and DSA-high subgroups. Both graft survival and CLAD-free survival were evaluated. Among factors that may contribute to DSA development we analyzed Pseudomonas aeruginosa (P. aeruginosa) infection in bronchoalveolar lavage (BAL) specimens. RESULTS: High MFI DSAs contributed to clinical antibody-mediated rejection (AMR) and were associated with significantly worse graft (HR: 5.77, p < 0.0001) and CLAD-free survival (HR: 6.47, p = 0.019) compared to low or negative MFI DSA levels. Analysis of BAL specimens revealed a strong correlation between DSA status, P. aeruginosa infection and BAL neutrophilia. DSA-high status and clinical AMR were both independent prognosticators for decreased graft and CLAD-free survival in our multivariate Cox-regression models, whereas BAL neutrophilia was associated with worse graft survival. CONCLUSIONS: P. aeruginosa infection rates are elevated in recipients with a strong DSA response. Our results indicate that the simultaneous interpretation of MFI values and BAL neutrophilia is a feasible approach for risk evaluation and may help clinicians when to initiate DSA desensitization therapy, as early intervention could improve prognosis.


Subject(s)
Graft Rejection , Lung Transplantation , Pseudomonas Infections , Pseudomonas aeruginosa , Lung Transplantation/adverse effects , Lung Transplantation/mortality , Humans , Female , Male , Middle Aged , Pseudomonas Infections/immunology , Pseudomonas Infections/diagnosis , Pseudomonas Infections/mortality , Adult , Pseudomonas aeruginosa/immunology , Graft Rejection/immunology , Graft Rejection/diagnosis , Tissue Donors , Retrospective Studies , Graft Survival , Cohort Studies , Isoantibodies/blood , Aged
7.
J Am Chem Soc ; 146(27): 18427-18439, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38946080

ABSTRACT

Pseudomonas aeruginosa bacteria are becoming increasingly resistant against multiple antibiotics. Therefore, the development of vaccines to prevent infections with these bacteria is an urgent medical need. While the immunological activity of lipopolysaccharide O-antigens in P. aeruginosa is well-known, the specific protective epitopes remain unidentified. Herein, we present the first chemical synthesis of highly functionalized aminoglycoside trisaccharide 1 and its acetamido derivative 2 found in the P. aeruginosa serotype O5 O-antigen. The synthesis of the trisaccharide targets is based on balancing the reactivity of disaccharide acceptors and monosaccharide donors. Glycosylations were analyzed by quantifying the reactivity of the hydroxyl group of the disaccharide acceptor using the orbital-weighted Fukui function and dual descriptor. The stereoselective formation of 1,2-cis-α-fucosylamine linkages was achieved through a combination of remote acyl participation and reagent modulation. The simultaneous SN2 substitution of azide groups at C2' and C2″ enabled the efficient synthesis of 1,2-cis-ß-linkages for both 2,3-diamino-D-mannuronic acids. Through a strategic orthogonal modification, the five amino groups on target trisaccharide 1 were equipped with a rare acetamidino (Am) and four acetyl (Ac) groups. Glycan microarray analyses of sera from patients infected with P. aeruginosa indicated that trisaccharides 1 and 2 are key antigenic epitopes of the serotype O5 O-antigen. The acetamidino group is not an essential determinant of antibody binding. The ß-D-ManpNAc3NAcA residue is a key motif for the antigenicity of serotype O5 O-antigen. These findings serve as a foundation for the development of glycoconjugate vaccines targeting P. aeruginosa serotype O5.


Subject(s)
Aminoglycosides , O Antigens , Pseudomonas aeruginosa , Trisaccharides , Pseudomonas aeruginosa/immunology , O Antigens/chemistry , O Antigens/immunology , Trisaccharides/chemistry , Trisaccharides/immunology , Trisaccharides/chemical synthesis , Aminoglycosides/chemistry , Aminoglycosides/chemical synthesis , Aminoglycosides/immunology
8.
Front Immunol ; 15: 1405364, 2024.
Article in English | MEDLINE | ID: mdl-39021568

ABSTRACT

Introduction: As the body's first line of defense against disease and infection, neutrophils must efficiently navigate to sites of inflammation; however, neutrophil dysregulation contributes to the pathogenesis of numerous diseases that leave people susceptible to infections. Many of these diseases are also associated with changes to the protein composition of the extracellular matrix. While it is known that neutrophils and endothelial cells, which play a key role in neutrophil activation, are sensitive to the mechanical and structural properties of the extracellular matrix, our understanding of how protein composition in the matrix affects the neutrophil response to infection is incomplete. Methods: To investigate the effects of extracellular matrix composition on the neutrophil response to infection, we used an infection-on-a-chip microfluidic device that replicates a portion of a blood vessel endothelium surrounded by a model extracellular matrix. Model blood vessels were fabricated by seeding human umbilical vein endothelial cells on 2, 4, or 6 mg/mL type I collagen hydrogels. Primary human neutrophils were loaded into the endothelial lumens and stimulated by adding the bacterial pathogen Pseudomonas aeruginosa to the surrounding matrix. Results: Collagen concentration did not affect the cell density or barrier function of the endothelial lumens. Upon infectious challenge, we found greater neutrophil extravasation into the 4 mg/mL collagen gels compared to the 6 mg/mL collagen gels. We further found that extravasated neutrophils had the highest migration speed and distance in 2mg/mL gels and that these values decreased with increasing collagen concentration. However, these phenomena were not observed in the absence of an endothelial lumen. Lastly, no differences in the percent of extravasated neutrophils producing reactive oxygen species were observed across the various collagen concentrations. Discussion: Our study suggests that neutrophil extravasation and migration in response to an infectious challenge are regulated by collagen concentration in an endothelial cell-dependent manner. The results demonstrate how the mechanical and structural aspects of the tissue microenvironment affect the neutrophil response to infection. Additionally, these findings underscore the importance of developing and using microphysiological systems for studying the regulatory factors that govern the neutrophil response.


Subject(s)
Cell Movement , Human Umbilical Vein Endothelial Cells , Neutrophils , Humans , Neutrophils/immunology , Neutrophils/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/physiology , Extracellular Matrix/metabolism , Collagen/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/immunology , Lab-On-A-Chip Devices , Neutrophil Activation , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Cells, Cultured
9.
Front Immunol ; 15: 1405376, 2024.
Article in English | MEDLINE | ID: mdl-39015565

ABSTRACT

Pseudomonas aeruginosa is a highly adaptable opportunistic pathogen capable of exploiting barriers and immune defects to cause chronic lung infections in conditions such as cystic fibrosis. In these contexts, host immune responses are ineffective at clearing persistent bacterial infection, instead driving a cycle of inflammatory lung damage. This review outlines key components of the host immune response to chronic P. aeruginosa infection within the lung, beginning with initial pathogen recognition, followed by a robust yet maladaptive innate immune response, and an ineffective adaptive immune response that propagates lung damage while permitting bacterial persistence. Untangling the interplay between host immunity and chronic P. aeruginosa infection will allow for the development and refinement of strategies to modulate immune-associated lung damage and potentiate the immune system to combat chronic infection more effectively.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Chronic Disease , Animals , Host-Pathogen Interactions/immunology , Adaptive Immunity , Lung Diseases/immunology , Lung Diseases/microbiology , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Lung/immunology , Lung/microbiology
10.
Int J Biol Macromol ; 277(Pt 1): 133964, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39029853

ABSTRACT

This study presents the design and fabrication of an innovative vaccine candidate targeting Pseudomonas aeruginosa (P. aeruginosa). The vaccine consists of gelatin nanoparticles (GNPs) encapsulating a chimeric protein (CP) derived from the ExoS and OprI proteins from P. aeruginosa. The physicochemical properties of the GNPs were assessed using dynamic light scattering (DLS) and electron microscopy. The toxicity, encapsulation efficacy, release profile, and effectiveness of CP-encapsulated GNPs (CP-GNPs) in an animal model were investigated. The resulting nanovaccine demonstrated uniform spherical particles with an average size of 135 nm and an encapsulation efficiency of 85 %. The release assay revealed that 23 % of the antigen was released from the CP-GNPs after 20 days. The GNPs did not exhibit any toxic effects on L929 cells in vitro. The formulation induced both systemic and mucosal antibody responses. Additionally, CP-GNPs stimulated cytokine responses, including IFN-γ, IL-4, and IL-17, indicating the induction of both humoral (Th2) and cellular (Th1) responses. The CP-encapsulated GNPs formulation effectively protected the mice lungs against experimental respiratory tract infection, reducing colony count and inflammation. These findings suggest that CP-GNPs hold promise as a potential strategy for preventing respiratory tract infections caused by P. aeruginosa. Further research is needed to explore its clinical application.


Subject(s)
Gelatin , Nanoparticles , Pseudomonas Infections , Pseudomonas aeruginosa , Respiratory Tract Infections , Animals , Gelatin/chemistry , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/drug effects , Nanoparticles/chemistry , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Mice , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/immunology , Cytokines/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/chemistry , Female , Adjuvants, Immunologic/pharmacology , Mice, Inbred BALB C , Cell Line
11.
Toxins (Basel) ; 16(6)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38922165

ABSTRACT

Extensively drug-resistant Pseudomonas aeruginosa infections are emerging as a significant threat associated with adverse patient outcomes. Due to this organism's inherent properties of developing antibiotic resistance, we sought to investigate alternative strategies such as identifying "high value" antigens for immunotherapy-based purposes. Through extensive database mining, we discovered that numerous Gram-negative bacterial (GNB) genomes, many of which are known multidrug-resistant (MDR) pathogens, including P. aeruginosa, horizontally acquired the evolutionarily conserved gene encoding Zonula occludens toxin (Zot) with a substantial degree of homology. The toxin's genomic footprint among so many different GNB stresses its evolutionary importance. By employing in silico techniques such as proteomic-based phylogenetic tracing, in conjunction with comparative structural modeling, we discovered a highly conserved intermembrane associated stretch of 70 amino acids shared among all the GNB strains analyzed. The characterization of our newly identified antigen reveals it to be a "high value" vaccine candidate specific for P. aeruginosa. This newly identified antigen harbors multiple non-overlapping B- and T-cell epitopes exhibiting very high binding affinities and can adopt identical tertiary structures among the least genetically homologous P. aeruginosa strains. Taken together, using proteomic-driven reverse vaccinology techniques, we identified multiple "high value" vaccine candidates capable of eliciting a polarized immune response against all the P. aeruginosa genetic variants tested.


Subject(s)
Phylogeny , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Humans , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/chemistry , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics
12.
Front Immunol ; 15: 1418061, 2024.
Article in English | MEDLINE | ID: mdl-38903499

ABSTRACT

Extracellular vesicles (EVs), characterized by low immunogenicity, high biocompatibility and targeting specificity along with excellent blood-brain barrier permeability, are increasingly recognized as promising drug delivery vehicles for treating a variety of diseases, such as cancer, inflammation and viral infection. However, recent findings demonstrate that the intracellular delivery efficiency of EVs fall short of expectations due to phagocytic clearance mediated by the host mononuclear phagocyte system through Fcγ receptors, complement receptors as well as non-opsonic phagocytic receptors. In this text, we investigate a range of bacterial virulence proteins that antagonize host phagocytic machinery, aiming to explore their potential in engineering EVs to counteract phagocytosis. Special emphasis is placed on IdeS secreted by Group A Streptococcus and ImpA secreted by Pseudomonas aeruginosa, as they not only counteract phagocytosis but also bind to highly upregulated surface biomarkers αVß3 on cancer cells or cleave the tumor growth and metastasis-promoting factor CD44, respectively. This suggests that bacterial anti-phagocytic proteins, after decorated onto EVs using pre-loading or post-loading strategies, can not only improve EV-based drug delivery efficiency by evading host phagocytosis and thus achieve better therapeutic outcomes but also further enable an innovative synergistic EV-based cancer therapy approach by integrating both phagocytosis antagonism and cancer targeting or deactivation.


Subject(s)
Extracellular Vesicles , Phagocytosis , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Phagocytosis/immunology , Humans , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/immunology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/immunology , Pseudomonas aeruginosa/immunology
13.
Front Immunol ; 15: 1398369, 2024.
Article in English | MEDLINE | ID: mdl-38835759

ABSTRACT

Introduction: Although many studies have underscored the importance of T cells, phenotypically and functionally, fewer have studied the functions of myeloid cells in COVID disease. In particular, the potential role of myeloid cells such as monocytes and low-density neutrophils (LDNs) in innate responses and particular in the defense against secondary bacterial infections has been much less documented. Methods: Here, we compared, in a longitudinal study, healthy subjects, idiopathic fibrosis patients, COVID patients who were either hospitalized/moderate (M-) or admitted to ICU (COV-ICU) and patients in ICU hospitalized for other reasons (non-COV-ICU). Results: We show that COVID patients have an increased proportion of low-density neutrophils (LDNs), which produce high levels of proteases (particularly, NE, MMP-8 and MMP-9) (unlike non-COV-ICU patients), which are partly responsible for causing type II alveolar cell damage in co-culture experiments. In addition, we showed that M- and ICU-COVID monocytes had reduced responsiveness towards further live Pseudomonas aeruginosa (PAO1 strain) infection, an important pathogen colonizing COVID patients in ICU, as assessed by an impaired secretion of myeloid cytokines (IL-1, TNF, IL-8,…). By contrast, lymphoid cytokines (in particular type 2/type 3) levels remained high, both basally and post PAO1 infection, as reflected by the unimpaired capacity of T cells to proliferate, when stimulated with anti-CD3/CD28 beads. Discussion: Overall, our results demonstrate that COVID circulatory T cells have a biased type 2/3 phenotype, unconducive to proper anti-viral responses and that myeloid cells have a dual deleterious phenotype, through their LDN-mediated damaging effect on alveolar cells and their impaired responsiveness (monocyte-mediated) towards bacterial pathogens such as P. aeruginosa.


Subject(s)
COVID-19 , Monocytes , Neutrophils , Pseudomonas Infections , Pseudomonas aeruginosa , SARS-CoV-2 , Humans , COVID-19/immunology , Pseudomonas aeruginosa/immunology , Monocytes/immunology , Male , Female , Middle Aged , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Pseudomonas Infections/immunology , Neutrophils/immunology , Aged , Cytokines/metabolism , Cytokines/immunology , Adult , Longitudinal Studies , Leukocytes, Mononuclear/immunology , Lung/immunology , Lung/pathology , Lung/microbiology
14.
PLoS Pathog ; 20(6): e1012252, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38833496

ABSTRACT

Microbial pathogenicity often depends on the route of infection. For instance, P. aeruginosa or S. marcescens cause acute systemic infections when low numbers of bacteria are injected into D. melanogaster flies whereas flies succumb much slower to the continuous ingestion of these pathogens, even though both manage to escape from the gut compartment and reach the hemocoel. Here, we have developed a latent P. aeruginosa infection model by feeding flies on the bacteria for a short period. The bacteria stably colonize internal tissues yet hardly cause any damage since latently-infected flies live almost as long as noninfected control flies. The apparently dormant bacteria display particular characteristics in terms of bacterial colony morphology, composition of the outer cell wall, and motility. The virulence of these bacteria can however be reactivated upon wounding the host. We show that melanization but not the cellular or the systemic humoral response is the predominant host defense that establishes latency and may coerce the bacteria to a dormant state. In addition, the lasting activation of the melanization responses in latently-infected flies provides a degree of protection to the host against a secondary fungal infection. Latent infection by an ingested pathogen protects against a variety of homologous or heterologous systemic secondary infectious challenges, a situation previously described for the endosymbiotic Wolbachia bacteria, a guard against viral infections.


Subject(s)
Drosophila melanogaster , Immunity, Innate , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Drosophila melanogaster/microbiology , Drosophila melanogaster/immunology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Virulence , Disease Models, Animal , Host-Pathogen Interactions/immunology
15.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38896653

ABSTRACT

Bacteria contain a wide variety of innate and adaptive immune systems which provide protection to the host against invading genetic material, including bacteriophages (phages). It is becoming increasingly clear that bacterial immune systems are frequently lost and gained through horizontal gene transfer. However, how and when new immune systems can become established in a bacterial population have remained largely unstudied. We developed a joint epidemiological and evolutionary model that predicts the conditions necessary for the spread of a CRISPR-Cas (clustered regularly interspaced short palindromic repeats-CRISPR-associated) immune system into a bacterial population lacking this system. We found that whether bacteria carrying CRISPR-Cas will spread (increase in frequency) into a bacterial population depends on the abundance of phages and the difference in the frequency of phage resistance mechanisms between bacteria carrying a CRISPR-Cas immune system and those not (denoted as ${f}_{\Delta }$). Specifically, the abundance of cells carrying CRISPR-Cas will increase if there is a higher proportion of phage resistance (either via CRISPR-Cas immunity or surface modification) in the CRISPR-Cas-possessing population than in the cells lacking CRISPR-Cas. We experimentally validated these predictions in a model using Pseudomonas aeruginosa PA14 and phage DMS3vir. Specifically, by varying the initial ratios of different strains of bacteria that carry alternative forms of phage resistance, we confirmed that the spread of cells carrying CRISPR-Cas through a population can be predicted based on phage density and the relative frequency of resistance phenotypes. Understanding which conditions promote the spread of CRISPR-Cas systems helps to predict when and where these defences can become established in bacterial populations after a horizontal gene transfer event, both in ecological and clinical contexts.


Subject(s)
Bacteria , Bacteriophages , CRISPR-Cas Systems , Bacteriophages/genetics , Bacteria/genetics , Bacteria/virology , Bacteria/classification , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Gene Transfer, Horizontal , Clustered Regularly Interspaced Short Palindromic Repeats
16.
J Immunol ; 213(3): 317-327, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38905107

ABSTRACT

Obesity is associated with increased morbidity and mortality during bacterial pneumonia. Cyclooxygenase-2 (COX-2) and PGE2 have been shown to be upregulated in patients who are obese. In this study, we investigated the role of obesity and PGE2 in bacterial pneumonia and how inhibition of PGE2 improves antibacterial functions of macrophages. C57BL/6J male and female mice were fed either a normal diet (ND) or high-fat diet (HFD) for 16 wk. After this time, animals were infected with Pseudomonas aeruginosa in the lung. In uninfected animals, alveolar macrophages were extracted for either RNA analysis or to be cultured ex vivo for functional analysis. HFD resulted in changes in immune cell numbers in both noninfected and infected animals. HFD animals had increased bacterial burden compared with ND animals; however, male HFD animals had higher bacterial burden compared with HFD females. Alveolar macrophages from HFD males had decreased ability to phagocytize and kill bacteria and were shown to have increased cyclooxygenase-2 and PGE2. Treating male, but not female, alveolar macrophages with PGE2 leads to increases in cAMP and decreased bacterial phagocytosis. Treatment with lumiracoxib-conjugated nanocarriers targeting alveolar macrophages improves bacterial phagocytosis and clearance in both ND and HFD male animals. Our study highlights that obesity leads to worse morbidity during bacterial pneumonia in male mice because of elevated PGE2. In addition, we uncover a sex difference in both obesity and infection, because females produce high basal PGE2 but because of a failure to signal via cAMP do not display impaired phagocytosis.


Subject(s)
Dinoprostone , Macrophages, Alveolar , Mice, Inbred C57BL , Obesity , Pneumonia, Bacterial , Pseudomonas Infections , Pseudomonas aeruginosa , Up-Regulation , Animals , Female , Male , Macrophages, Alveolar/immunology , Mice , Dinoprostone/metabolism , Pseudomonas aeruginosa/immunology , Obesity/immunology , Pseudomonas Infections/immunology , Pneumonia, Bacterial/immunology , Up-Regulation/immunology , Diet, High-Fat/adverse effects , Cyclooxygenase 2/metabolism , Phagocytosis/immunology , Sex Factors
17.
J Immunol Methods ; 531: 113701, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852836

ABSTRACT

Flagellum-mediated motility is essential to Pseudomonas aeruginosa (P. aeruginosa) virulence. Antibody against flagellin reduces motility and inhibits the spread of the bacteria from the infection site. The standard soft-agar assay to demonstrate anti-flagella motility inhibition requires long incubation times, is difficult to interpret, and requires large amounts of antibody. We have developed a time-lapse video microscopy method to analyze anti-flagellin P. aeruginosa motility inhibition that has several advantages over the soft agar assay. Antisera from mice immunized with flagellin type A or B were incubated with Green Fluorescent Protein (GFP)-expressing P. aeruginosa strain PAO1 (FlaB+) and GFP-expressing P. aeruginosa strain PAK (FlaA+). We analyzed the motion of the bacteria in video taken in ten second time intervals. An easily measurable decrease in bacterial locomotion was observed microscopically within minutes after the addition of small volumes of flagellin antiserum. From data analysis, we were able to quantify the efficacy of anti-flagellin antibodies in the test serum that decreased P. aeruginosa motility. This new video microscopy method to assess functional activity of anti-flagellin antibodies required less serum, less time, and had more robust and reproducible endpoints than the standard soft agar motility inhibition assay.


Subject(s)
Antibodies, Bacterial , Flagella , Flagellin , Immune Sera , Microscopy, Video , Pseudomonas aeruginosa , Flagellin/immunology , Pseudomonas aeruginosa/immunology , Animals , Immune Sera/immunology , Antibodies, Bacterial/immunology , Flagella/immunology , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology
18.
Nanoscale ; 16(21): 10306-10317, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38727538

ABSTRACT

As a highly contagious opportunistic pathogen, Pseudomonas aeruginosa (P. aeruginosa) is one of the main causes of healthcare-associated infections. The drug-resistant nature of P. aeruginosa can render antibiotic treatments ineffective, leading to a high morbidity and mortality. Higher specificity and reduced toxicity are features of immunotherapy, which can generate robust immune responses and preserve long-term immunological memory to completely eradicate infections. In this study, we developed a type of P. aeruginosa vaccine based on a metal-organic framework. Specifically, MIL-101-Al nanoparticles were synthesized to encapsulate antigens derived from the bacterial lysate (BL) of PAO1, a drug-resistant P. aeruginosa, and the adjuvant unmethylated cytosine-phosphate-guanine oligonucleotide (CpG), which were then modified with palmitic acid (PAA) to obtain MIL-BC@PAA. The stability and biocompatibility were significantly increased by capping with PAA. Moreover, MIL-BC@PAA showed significantly enhanced uptake by antigen presenting cells (APCs), and promoted their maturation. Importantly, immunity studies revealed the greatly elicited antigen-specific humoral and cellular responses, and a protection rate of about 70% was observed in P. aeruginosa-challenged mice. Overall, these results demonstrate the promising potential of MIL-BC@PAA as an ideal nanovaccine for P. aeruginosa vaccination.


Subject(s)
Adjuvants, Immunologic , Metal-Organic Frameworks , Palmitic Acid , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/drug effects , Animals , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/prevention & control , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Palmitic Acid/chemistry , Female , Nanoparticles/chemistry , Mice, Inbred BALB C , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacology
19.
STAR Protoc ; 5(2): 103070, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38768031

ABSTRACT

The nematode Caenorhabditis elegans is a powerful model organism for studying the molecular and cellular mechanisms of innate immunity governed by the intestine. Here, we present a protocol to perform C. elegans survival assays to infection by the bacterial pathogen Pseudomonas aeruginosa PA14. Specifically, we describe steps for preparing C. elegans strains and PA14 bacteria for survival assays. This protocol will assist researchers to study genes involved in intestinal innate immunity and gut defense against pathogen infection. For complete details on the use and execution of this protocol, please refer to Liu et al.1 and Zheng et al.2.


Subject(s)
Caenorhabditis elegans , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/immunology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Immunity, Innate
20.
Front Immunol ; 15: 1372349, 2024.
Article in English | MEDLINE | ID: mdl-38698863

ABSTRACT

Pseudomonas aeruginosa (Pa) is an opportunistic bacterial pathogen responsible for severe hospital acquired infections in immunocompromised and elderly individuals. Emergence of increasingly drug resistant strains and the absence of a broad-spectrum prophylactic vaccine against both T3SA+ (type III secretion apparatus) and ExlA+/T3SA- Pa strains worsen the situation in a post-pandemic world. Thus, we formulated a candidate subunit vaccine (called ExlA/L-PaF/BECC/ME) against both Pa types. This bivalent vaccine was generated by combining the C-terminal active moiety of exolysin A (ExlA) produced by non-T3SA Pa strains with our T3SA-based vaccine platform, L-PaF, in an oil-in-water emulsion. The ExlA/L-PaF in ME (MedImmune emulsion) was then mixed with BECC438b, an engineered lipid A analogue and a TLR4 agonist. This formulation was administered intranasally (IN) to young and elderly mice to determine its potency across a diverse age-range. The elderly mice were used to mimic the infection seen in elderly humans, who are more susceptible to serious Pa disease compared to their young adult counterparts. After Pa infection, mice immunized with ExlA/L-PaF/BECC/ME displayed a T cell-mediated adaptive response while PBS-vaccinated mice experienced a rapid onset inflammatory response. Important genes and pathways were observed, which give rise to an anti-Pa immune response. Thus, this vaccine has the potential to protect aged individuals in our population from serious Pa infection.


Subject(s)
Emulsions , Pseudomonas Infections , Pseudomonas Vaccines , Pseudomonas aeruginosa , Vaccines, Subunit , Animals , Pseudomonas aeruginosa/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/administration & dosage , Female , Vaccine Development , Humans , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Disease Models, Animal , Bacterial Proteins/immunology , Bacterial Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL