Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.560
Filter
1.
Immunohorizons ; 8(9): 606-621, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39240270

ABSTRACT

The human body harbors a substantial population of bacteria, which may outnumber host cells. Thus, there are multiple interactions between both cell types. Given the common presence of Staphylococcus aureus in the human body and the role of Th17 cells in controlling this pathogen on mucous membranes, we sought to investigate the effect of α-hemolysin, which is produced by this bacterium, on differentiating Th17 cells. RNA sequencing analysis revealed that α-hemolysin influences the expression of signature genes for Th17 cells as well as genes involved in epigenetic regulation. We observed alterations in various histone marks and genome methylation levels via whole-genome bisulfite sequencing. Our findings underscore how bacterial proteins can significantly influence the transcriptome, epigenome, and phenotype of human Th17 cells, highlighting the intricate and complex nature of the interaction between immune cells and the microbiota.


Subject(s)
Bacterial Toxins , Epigenesis, Genetic , Hemolysin Proteins , Staphylococcus aureus , Th17 Cells , Staphylococcus aureus/immunology , Staphylococcus aureus/genetics , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Hemolysin Proteins/metabolism , Humans , Th17 Cells/immunology , Th17 Cells/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , DNA Methylation , Cell Differentiation , Transcriptome
2.
Bull Exp Biol Med ; 177(2): 243-247, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39090462

ABSTRACT

The influence of non-opsonized and opsonized S. aureus 2879M and E. coli 321 strains on the total strength of interaction between the endothelial cell and neutrophil during the docking process was studied using in vitro model of experimental septicemia. We observed a decrease in the force and work of adhesion between receptors of neutrophils and endothelial cells under the influence of non-opsonized strains and further decrease in the affinity of single interactions between cells under the influence of opsonized S. aureus, which was compensated by an increase in the number of contacts, as well as an increase in the force of adhesion under the influence of opsonized E. coli compared to non-opsonized bacteria, which remained below the control level, while adhesion work reaches the control level. Thus, opsonization of S. aureus aggravates the "immunological uncoupling" between neutrophils and endothelial cells, while opsonization of E. coli reduces the pathological effect compared to non-opsonized bacteria.


Subject(s)
Endothelial Cells , Escherichia coli , Neutrophils , Sepsis , Staphylococcus aureus , Neutrophils/immunology , Neutrophils/metabolism , Escherichia coli/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Sepsis/immunology , Sepsis/microbiology , Sepsis/metabolism , Sepsis/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Humans , Phagocytosis , Cell Adhesion/immunology , Opsonin Proteins/metabolism , Opsonin Proteins/immunology , Bacterial Adhesion , Animals
3.
Front Immunol ; 15: 1397330, 2024.
Article in English | MEDLINE | ID: mdl-39185412

ABSTRACT

Introduction: The role of suppressor of cytokine signaling (SOCS)2 in anti-infective bacterial immunity has been poorly investigated compared to other members of the SOCS family. Methods: We characterized the previously identified loss of function R96C point mutation of SOCS2 using a genome-edited mouse model that resumes the phenotype of Socs2 knockout mice. The response of macrophages to TLR-ligands and Staphylococcus aureus was examined. Results and discussion: Conversely to previously published data using human monocyte-derived macrophages, the stimulation of bone-marrow-derived macrophages with various TLR ligands did not show any difference according to the SOCS2 variant. Upregulation of IL-6 and TNF-α pro-inflammatory cytokines production was only seen when the SOCS2 expression was promoted by the culture of macrophages in the presence of GM-CSF. Furthermore, we showed that the SOCS2 point mutation is associated with heightened STAT5 phosphorylation in a short time frame upon GM-CSF incubation. In mice, recruitment of neutrophil and F4/80int Ly6C+ inflammatory macrophage, as well as IFN-γ and IL-10 concentrations, are significantly increased upon S. aureus peritoneal infection. Altogether, these data support the idea that by lowering the pro-inflammatory environment, SOCS2 favors better control of bacterial burden during a systemic infection caused by S. aureus.


Subject(s)
Macrophages , Mice, Knockout , Staphylococcal Infections , Staphylococcus aureus , Suppressor of Cytokine Signaling Proteins , Animals , Staphylococcus aureus/immunology , Mice , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/genetics , Toll-Like Receptors/metabolism , Toll-Like Receptors/genetics , Cytokines/metabolism , Loss of Function Mutation , Inflammation/immunology , Inflammation/genetics , Mice, Inbred C57BL , Ligands , Humans
4.
PLoS Pathog ; 20(8): e1012437, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102432

ABSTRACT

The ability of Staphylococcus aureus (S. aureus) to survive within macrophages is a critical strategy for immune evasion, contributing to the pathogenesis and progression of osteomyelitis. However, the underlying mechanisms remain poorly characterized. This study discovered that inhibiting the MEK1/2 pathway reduced bacterial load and mitigated bone destruction in a mouse model of S. aureus osteomyelitis. Histological staining revealed increased phosphorylated MEK1/2 levels in bone marrow macrophages surrounding abscess in the mouse model of S. aureus osteomyelitis. Activation of MEK1/2 pathway and its roles in impairing macrophage bactericidal function were confirmed in primary mouse bone marrow-derived macrophages (BMDMs). Transcriptome analysis and in vitro experiments demonstrated that S. aureus activates the MEK1/2 pathway through EGFR signaling. Moreover, we found that excessive activation of EGFR-MEK1/2 cascade downregulates mitochondrial reactive oxygen species (mtROS) levels by suppressing Chek2 expression, thereby impairing macrophage bactericidal function. Furthermore, pharmacological inhibition of EGFR signaling prevented upregulation of phosphorylated MEK1/2 and restored Chek2 expression in macrophages, significantly enhancing S. aureus clearance and improving bone microstructure in vivo. These findings highlight the critical role of the EGFR-MEK1/2 cascade in host immune defense against S. aureus, suggesting that S. aureus may reduce mtROS levels by overactivating the EGFR-MEK1/2 cascade, thereby suppressing macrophage bactericidal function. Therefore, combining EGFR-MEK1/2 pathway blockade with antibiotics could represent an effective therapeutic approach for the treatment of S. aureus osteomyelitis.


Subject(s)
ErbB Receptors , MAP Kinase Kinase 1 , Macrophages , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Osteomyelitis/microbiology , Osteomyelitis/immunology , Osteomyelitis/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Mice , Staphylococcus aureus/immunology , ErbB Receptors/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL , Disease Models, Animal , Signal Transduction
5.
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
6.
J Mol Graph Model ; 132: 108848, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39182254

ABSTRACT

Staphylococcus aureus is a common bacterium that causes a variety of infections in humans. This microorganism produces several virulence factors, including hemolysins, which contribute to its disease-causing ability. The treatment of S. aureus infections typically involves the use of antibiotics. However, the emergence of antibiotic-resistant strains has become a major concern. Therefore, vaccination against S. aureus has gained attention as an alternative approach. Vaccination has the advantage of stimulating the immune system to produce specific antibodies that can neutralize bacteria and prevent infection. However, developing an effective vaccine against S. aureus has proven to be challenging. This study aimed to use in silico methods to design a multi-epitope vaccine against S. aureus infection based on hemolysin proteins. The designed vaccine contained four B-cell epitopes, four CTL epitopes, and four HTL epitopes, as well as the ribosomal protein L7/L12 and pan-HLA DR-binding epitope, included as adjuvants. Furthermore, the vaccine was non-allergenic and non-toxic with the potential to stimulate the TLR2-, TLR-4, and TLR-6 receptors. The predicted vaccine exhibited a high degree of antigenicity and stability, suggesting potential for further development as a viable vaccine candidate. The population coverage of the vaccine was 94.4 %, indicating potential widespread protection against S. aureus. Overall, these findings provide valuable insights into the design of an effective multi-epitope vaccine against S. aureus infection and pave the way for future experimental validations.


Subject(s)
Epitopes, B-Lymphocyte , Hemolysin Proteins , Staphylococcus aureus , Hemolysin Proteins/immunology , Hemolysin Proteins/chemistry , Staphylococcus aureus/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Humans , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/chemistry , Computational Biology/methods , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Molecular Docking Simulation , Epitopes/immunology , Epitopes/chemistry , Amino Acid Sequence
7.
Proc Natl Acad Sci U S A ; 121(36): e2321939121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39186649

ABSTRACT

Developing an effective Staphylococcus aureus (S. aureus) vaccine has been a challenging endeavor, as demonstrated by numerous failed clinical trials over the years. In this study, we formulated a vaccine containing a highly conserved moonlighting protein, the pyruvate dehydrogenase complex E2 subunit (PDHC), and showed that it induced strong protective immunity against epidemiologically relevant staphylococcal strains in various murine disease models. While antibody responses contributed to bacterial control, they were not essential for protective immunity in the bloodstream infection model. Conversely, vaccine-induced systemic immunity relied on γδ T cells. It has been suggested that prior S. aureus exposure may contribute to the reduction of vaccine efficacy. However, PDHC-induced protective immunity still facilitated bacterial clearance in mice previously exposed to S. aureus. Collectively, our findings indicate that PDHC is a promising serotype-independent vaccine candidate effective against both methicillin-sensitive and methicillin-resistant S. aureus isolates.


Subject(s)
Staphylococcal Infections , Staphylococcal Vaccines , Staphylococcus aureus , Animals , Staphylococcal Infections/prevention & control , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Mice , Staphylococcus aureus/immunology , Staphylococcus aureus/enzymology , Staphylococcal Vaccines/immunology , Pyruvate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex/immunology , Female , Antibodies, Bacterial/immunology , Disease Models, Animal , Humans , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Mice, Inbred C57BL , Methicillin-Resistant Staphylococcus aureus/immunology , Pyruvate Dehydrogenase (Lipoamide)/immunology , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase (Lipoamide)/genetics
9.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39150449

ABSTRACT

The pursuit of a vaccine to quell Staphylococcus aureus disease has been unfruitful. In this Viewpoint, we explore the biological linkage between microbial niche acquisition and host immunity as a basis to guide future vaccine efforts.


Subject(s)
Staphylococcal Infections , Staphylococcal Vaccines , Staphylococcus aureus , Vaccine Development , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Humans , Staphylococcal Infections/prevention & control , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Animals
10.
Vaccine ; 42(22): 126204, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39126830

ABSTRACT

The ESKAPE family, comprising Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp., poses a significant global threat due to their heightened virulence and extensive antibiotic resistance. These pathogens contribute largely to the prevalence of nosocomial or hospital-acquired infections, resulting in high morbidity and mortality rates. To tackle this healthcare problem urgent measures are needed, including development of innovative vaccines and therapeutic strategies. Designing vaccines involves a complex and resource-intensive process of identifying protective antigens and potential vaccine candidates (PVCs) from pathogens. Reverse vaccinology (RV), an approach based on genomics, made this process more efficient by leveraging bioinformatics tools to identify potential vaccine candidates. In recent years, artificial intelligence and machine learning (ML) techniques has shown promise in enhancing the accuracy and efficiency of reverse vaccinology. This study introduces a supervised ML classification framework, to predict potential vaccine candidates specifically against ESKAPE pathogens. The model's training utilized biological and physicochemical properties from a dataset containing protective antigens and non-protective proteins of ESKAPE pathogens. Conventional autoencoders based strategy was employed for feature encoding and selection. During the training process, seven machine learning algorithms were trained and subjected to Stratified 5-fold Cross Validation. Random Forest and Logistic Regression exhibited best performance in various metrics including accuracy, precision, recall, WF1 score, and Area under the curve. An ensemble model was developed, to take collective strengths of both the algorithms. To assess efficacy of our final ensemble model, a high-quality benchmark dataset was employed. VacSol-ML(ESKAPE) demonstrated outstanding discrimination between protective vaccine candidates (PVCs) and non-protective antigens. VacSol-ML(ESKAPE), proves to be an invaluable tool in expediting vaccine development for these pathogens. Accessible to the public through both a web server and standalone version, it encourages collaborative research. The web-based and standalone tools are available at http://vacsolml.mgbio.tech/.


Subject(s)
Antigens, Bacterial , Bacterial Vaccines , Machine Learning , Antigens, Bacterial/immunology , Humans , Bacterial Vaccines/immunology , Klebsiella pneumoniae/immunology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Enterococcus faecium/immunology , Enterococcus faecium/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/genetics , Acinetobacter baumannii/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Computational Biology/methods , Enterobacter/immunology , Enterobacter/genetics , Vaccinology/methods
11.
Int Immunopharmacol ; 140: 112924, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39133958

ABSTRACT

Staphylococcus aureus (SA) is a common Gram-positive bacterium that activates inflammatory cells, expressing various cytokines and inducing an inflammatory response. Recent research revealed aconitate decarboxylase 1 (ACOD1) as a regulator of the immune response through various metabolic pathways, playing a dual role in the inflammatory response. However, the mechanism by which ACOD1 participates in the regulation of SA-induced inflammatory responses in macrophages remains unknown. Therefore, this study aims to investigate the function and underlying regulatory mechanisms of ACOD1 in SA-induced inflammatory response. This study reveals that SA induced a macrophage inflammatory response and upregulated ACOD1 expression. ACOD1 knockdown significantly inhibited SA-induced macrophage inflammatory response, attenuated SA-induced nuclear envelope wrinkling, and plasma membrane rupture, and suppressed the TLR4/NF-κB signaling pathway. Furthermore, ACOD1 knockdown reduced the inflammatory response and alleviated lung tissue injury and cellular damage, leading to decreased bacterial loads in the lungs of SA-infected mice. Collectively, these findings demonstrate that SA induces an inflammatory response in macrophages and increases ACOD1 expression. ACOD1 enhances SA-induced inflammatory responses via the TLR4/NF-κB signaling pathway. Our findings highlight the significant role of ACOD1 in mediating the inflammatory response in SA-infected macrophages and elucidate its molecular mechanism in regulating the SA-induced inflammatory response.


Subject(s)
Carboxy-Lyases , Macrophages , Signal Transduction , Staphylococcal Infections , Staphylococcus aureus , Animals , Humans , Mice , Carboxy-Lyases/metabolism , Carboxy-Lyases/genetics , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/pathology , Lung/microbiology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics
12.
Adv Exp Med Biol ; 1445: 151-156, 2024.
Article in English | MEDLINE | ID: mdl-38967757

ABSTRACT

Skin is the most prominent tissue and organ, as well as the first line of defence, of the body. Because it is situated on the body's surface, it is constantly exposed to microbial, chemical, and physical factors such as mechanical stimulation. Therefore, skin has evolved substantial immune defences, regenerative ability, and anti-injury capacity. Epidermal cells produce antibacterial peptides that play a role in immune defence under physiological conditions. Additionally, IgG or IgA in the skin also participates in local anti-infective immunity. However, based on the classical theory of immunology, Ig can only be produced by B cells which should be derived from local B cells. This year, thanks to the discovery of Ig derived from non B cells (non B-Ig), Ig has also been found to be expressed in epidermal cells and contributes to immune defence. Epidermal cell-derived IgG and IgA have been demonstrated to have potential antibody activity by binding to pathogens. However, these epidermal cell-derived Igs show different microbial binding characteristics. For instance, IgG binds to Staphylococcus aureus and IgA binds to Staphylococcus epidermidis. Epidermal cells producing IgG and IgA may serve as an effective defense mechanism alongside B cells, providing a novel insight into skin immunity.


Subject(s)
Immunoglobulin A , Skin , Humans , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Skin/immunology , Animals , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , B-Lymphocytes/immunology , Immunoglobulins/immunology , Immunoglobulins/metabolism , Staphylococcus aureus/immunology , Staphylococcus epidermidis/immunology , Epidermis/immunology , Epidermis/metabolism , Epidermal Cells/immunology , Epidermal Cells/metabolism
13.
JCI Insight ; 9(13)2024 May 28.
Article in English | MEDLINE | ID: mdl-38973612

ABSTRACT

Staphylococcus aureus is a major human pathogen. An effective anti-S. aureus vaccine remains elusive as the correlates of protection are ill-defined. Targeting specific T cell populations is an important strategy for improving anti-S. aureus vaccine efficacy. Potential bottlenecks that remain are S. aureus-induced immunosuppression and the impact this might have on vaccine-induced immunity. S. aureus induces IL-10, which impedes effector T cell responses, facilitating persistence during both colonization and infection. Thus, it was hypothesized that transient targeting of IL-10 might represent an innovative way to improve vaccine efficacy. In this study, IL-10 expression was elevated in the nares of persistent carriers of S. aureus, and this was associated with reduced systemic S. aureus-specific Th1 responses. This suggests that systemic responses are remodeled because of commensal exposure to S. aureus, which negatively implicates vaccine function. To provide proof of concept that targeting immunosuppressive responses during immunization may be a useful approach to improve vaccine efficacy, we immunized mice with T cell-activating vaccines in combination with IL-10-neutralizing antibodies. Blocking IL-10 during vaccination enhanced effector T cell responses and improved bacterial clearance during subsequent systemic and subcutaneous infection. Taken together, these results reveal a potentially novel strategy for improving anti-S. aureus vaccine efficacy.


Subject(s)
Interleukin-10 , Staphylococcal Infections , Staphylococcal Vaccines , Staphylococcus aureus , Interleukin-10/metabolism , Interleukin-10/immunology , Animals , Staphylococcal Infections/prevention & control , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Mice , Staphylococcus aureus/immunology , Female , Mice, Inbred C57BL , Th1 Cells/immunology , Immunization/methods , Humans , Antibodies, Neutralizing/immunology , Vaccine Efficacy , Vaccination/methods
14.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000557

ABSTRACT

The effects of intestinal microflora on extraintestinal immune response by intestinal cytokines and metabolites have been documented, but whether intestinal microbes stimulate serum antibody generation is unknown. Here, serum antibodies against 69 outer membrane proteins of Escherichia coli, a dominant bacterium in the human intestine, are detected in 141 healthy individuals of varying ages. Antibodies against E. coli outer membrane proteins are determined in all serum samples tested, and frequencies of antibodies to five outer membrane proteins (OmpA, OmpX, TsX, HlpA, and FepA) are close to 100%. Serum antibodies against E. coli outer membrane proteins are further validated by Western blot and bacterial pull-down. Moreover, the present study shows that OstA, HlpA, Tsx, NlpB, OmpC, YfcU, and OmpA provide specific immune protection against pathogenic E. coli, while HlpA and OmpA also exhibit cross-protection against Staphylococcus aureus infection. These finding indicate that intestinal E. coli activate extraintestinal antibody responses and provide anti-infective immunity.


Subject(s)
Antibodies, Bacterial , Bacterial Outer Membrane Proteins , Escherichia coli , Humans , Escherichia coli/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/immunology , Adult , Female , Staphylococcus aureus/immunology , Male , Antibody Formation/immunology , Middle Aged , Escherichia coli Proteins/immunology , Young Adult , Aged , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Adolescent , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology
15.
Sci Rep ; 14(1): 15472, 2024 07 05.
Article in English | MEDLINE | ID: mdl-38969796

ABSTRACT

This study evaluated the determinants of mortality and the T cell immune response in patients with persistent Staphylococcus aureus bacteremia (SAB). This was a prospective cohort study and patients with confirmed SAB were enrolled from 2008 to 2020. We compared clinical, microbiological, and genotypic features between surviving and deceased patients with persistent SAB. The concentrations of cytokines and the proportions of IFN-γ secreting CD4+ T cells were measured serially during the bacteremia period. Of the 1760 patients, 242 had persistent bacteremia (PB), and 49 PB patients died within 30 days. In the multivariate analysis, the APACHE II score and female sex were independently associated with 30 days mortality. The level of IL-10 was significantly increased in the plasma of patients with a high Pitt bacteremia score and those who died within 12 weeks from the index day. The proportion of IFN-γ-secreting CD4+ T cells were the highest just before the positive-to-negative conversion of blood cultures in patients with a low Pitt bacteremia score and those who survived for 12 weeks. The level of IL-10 is correlated with clinical outcomes in PB patients. IFN-γ secreting CD4+ T cells might play a pivotal role in SAB PB.


Subject(s)
Bacteremia , CD4-Positive T-Lymphocytes , Staphylococcal Infections , Staphylococcus aureus , Humans , Male , Female , Bacteremia/mortality , Bacteremia/microbiology , Bacteremia/immunology , CD4-Positive T-Lymphocytes/immunology , Staphylococcal Infections/mortality , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology , Middle Aged , Risk Factors , Aged , Prospective Studies , Interferon-gamma/blood , Interferon-gamma/metabolism , Interleukin-10/blood , Adult , Cytokines/blood , Cytokines/metabolism
16.
Int J Biol Macromol ; 276(Pt 2): 133957, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39029852

ABSTRACT

Staphylococcal Enterotoxin Type B (SEB), produced by Staphylococcus aureus bacteria, is notorious for inducing severe food poisoning and toxic shock syndrome. While nanobody-based treatments hold promises for combating SEB-induced diseases, the lack of structural information between SEB and nanobodies has hindered the development of nanobody-based therapeutics. Here, we present crystal structures of SEB-Nb3, SEB-Nb6, SEB-Nb8, SEB-Nb11, and SEB-Nb20 at resolutions ranging from 1.59 Å to 2.33 Å. Crystallographic analysis revealed that Nb3, Nb8, Nb11, and Nb20 bind to SEB at the T-cell receptor (TCR) interface, while Nb6 binds at the major histocompatibility complex (MHC) interface, suggesting their potential to inhibit SEB function by disrupting interactions with TCR or MHC molecules. Molecular biological analyses confirmed the thermodynamic and kinetic parameters of Nb3, Nb5, Nb6, Nb8, Nb11, Nb15, Nb18, and Nb20 to SEB. The competitive inhibition was further confirmed by cell-based experiments demonstrating nanobody neutralization. These findings elucidate the structural basis for developing specific nanobodies to neutralize SEB threats, providing crucial insights into the underlying mechanisms and offering significant assistance for further optimization towards future therapeutic strategies.


Subject(s)
Enterotoxins , Protein Binding , Single-Domain Antibodies , Enterotoxins/chemistry , Enterotoxins/immunology , Enterotoxins/metabolism , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Humans , Models, Molecular , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/chemistry , Staphylococcus aureus/immunology , Crystallography, X-Ray , Thermodynamics , Kinetics
17.
Cell Mol Life Sci ; 81(1): 300, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39001897

ABSTRACT

BACKGROUND: Age-associated impairments in innate immunity are believed to be a causative factor responsible for severe pathogenesis of Staphylococcus aureus (S. aureus) infection in the bone tissue. However, the basis for age-associated decline in innate immune response upon S. aureus infection remains poorly understood. RESULTS: Our transcriptional data (GEO: GSE166522) from a mouse model of S. aureus osteomyelitis show up-regulated CXCL9 and CXCL10 (CXCL9/10), which is further confirmed in vitro and in vivo by the present study. Notably, monocytes are a main source for CXCL9/10 production in bone marrow upon S. aureus challenge, but this response declines in middle-aged mice. Interestingly, conditional medium of bone marrow monocytes from middle-aged mice has a strikingly decreased effect on bactericidal functions of neutrophils and macrophages compares with that from young mice. We further show that activation of CXCL9/10-CXCR3 axis between monocytes and macrophages/neutrophils promotes the bactericidal function of the cells, whereas blocking the axis impairs such function. Importantly, treatment with either exogenous CXCL9 or CXCL10 in a middle-aged mice model enhances, while pharmacological inhibition of CXCR3 in young mice model impairs, bacterial clearance and bone marrow structure. CONCLUSIONS: These findings demonstrate that bone marrow monocytes act as a critical promotor of innate immune response via the CXLCL9/10-CXCR3 axis upon S. aureus infection, and that the increased susceptibility to S. aureus infection in skeleton in an aged host may be largely attributable to the declined induction of CXCR9/10 in monocytes.


Subject(s)
Chemokine CXCL10 , Chemokine CXCL9 , Disease Models, Animal , Immunity, Innate , Monocytes , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Osteomyelitis/microbiology , Osteomyelitis/immunology , Osteomyelitis/metabolism , Osteomyelitis/pathology , Monocytes/immunology , Monocytes/metabolism , Chemokine CXCL9/metabolism , Chemokine CXCL9/genetics , Staphylococcus aureus/immunology , Mice , Chemokine CXCL10/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcal Infections/metabolism , Mice, Inbred C57BL , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Aging/immunology , Neutrophils/immunology , Neutrophils/metabolism , Macrophages/immunology , Macrophages/metabolism
18.
BMC Microbiol ; 24(1): 280, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068414

ABSTRACT

BACKGROUND: Enterococcus faecium and Staphylococcus aureus are the Gram-positive pathogens of the ESKAPE group, known to represent a great threat to human health due to their high virulence and multiple resistances to antibiotics. Combined, enterococci and S. aureus account for 26% of healthcare-associated infections and are the most common organisms responsible for blood stream infections. We previously showed that the peptidyl-prolyl cis/trans isomerase (PPIase) PpiC of E. faecium elicits the production of specific, opsonic, and protective antibodies that are effective against several strains of E. faecium and E. faecalis. Due to the ubiquitous characteristics of PPIases and their essential function within Gram-positive cells, we hypothesized a potential cross-reactive effect of anti-PpiC antibodies. RESULTS: Opsonophagocytic assays combined with bioinformatics led to the identification of the foldase protein PrsA as a new potential vaccine antigen in S. aureus. We show that PrsA is a stable dimeric protein able to elicit opsonic antibodies against the S. aureus strain MW2, as well as cross-binding and cross-opsonic in several S. aureus, E. faecium and E. faecalis strains. CONCLUSIONS: Given the multiple antibiotic resistances S. aureus and enterococci present, finding preventive strategies is essential to fight those two nosocomial pathogens. The study shows the potential of PrsA as an antigen to use in vaccine formulation against the two dangerous Gram-positive ESKAPE bacteria. Our findings support the idea that PPIases should be further investigated as vaccine targets in the frame of pan-vaccinomics strategy.


Subject(s)
Bacterial Proteins , Enterococcus faecalis , Enterococcus faecium , Peptidylprolyl Isomerase , Staphylococcus aureus , Staphylococcus aureus/immunology , Staphylococcus aureus/genetics , Enterococcus faecium/immunology , Enterococcus faecium/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Peptidylprolyl Isomerase/immunology , Peptidylprolyl Isomerase/genetics , Enterococcus faecalis/immunology , Enterococcus faecalis/genetics , Humans , Gram-Positive Bacterial Infections/prevention & control , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/microbiology , Bacterial Vaccines/immunology , Opsonin Proteins/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Animals , Cross Reactions , Mice , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Phagocytosis , Staphylococcal Infections/prevention & control , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology
19.
Arch Microbiol ; 206(8): 361, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066807

ABSTRACT

In the complex realm of bacterial infections, particularly those caused by Staphylococcus aureus (S. aureus), macrophages play a pivotal role in orchestrating the immune response. During the initial stages of infection, the monocytes give rise to macrophages with a pro-inflammatory (M1 type) behaviour, engulfing and neutralizing the invading pathogens. However, under the sustained influence of S. aureus infection, monocytes can undergo a transition into an anti-inflammatory M2 state (pro-infection) rather than the M1 state (anti-infection), thereby compromising effective infection control. Therefore, it is necessary to develop a strategy that would preserve the pro-inflammatory functions of macrophages, in a safe and controlled manner. For this, we focused on harnessing the potential of S. aureus-derived ghost cells (GCs) which are non-live empty envelopes of bacterial cells, but with the antigenic determinants intact. Through a unique Lugol's-iodine treatment, we generated GCs and characterization of these GCs using gel electrophoresis, FTIR, flow cytometry, TEM, and SEM confirmed their structural integrity. Following this, we assessed the extend of cellular association of the GCs with RAW267.4 macrophages, and observed an immediate interaction between the two, as evident from the flowcytometry and microscopy studies. We then performed macrophage polarisation on a human monocyte-macrophage model cell line, THP-1. Our findings revealed that GCs effectively activated macrophages, and promoted a pro-inflammatory polarisation with the expression of M1 differentiation markers (CD86, TNFα, IL-1ß, IL-6, IL-12) evaluated through both qPCR and ELISA. Interestingly an intermediary expression of M2 markers viz., CD206 and IL-10 was also observed, but was overruled by the enhanced expression of M1 markers at a later time point. Overall, our study introduces a novel approach utilizing GCs to guide naïve macrophages towards M1 subtypes, thereby potentiating immune responses during microbial infections. This innovative strategy can modulate macrophage function, ultimately improving outcomes in S. aureus infections and beyond.


Subject(s)
Cell Differentiation , Macrophages , Staphylococcal Infections , Staphylococcus aureus , Macrophages/immunology , Macrophages/microbiology , Staphylococcus aureus/immunology , Humans , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Animals , Mice , Monocytes/immunology , Monocytes/microbiology , Cytokines/metabolism , Cytokines/immunology , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism , Inflammation/immunology
20.
Talanta ; 278: 126525, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38991406

ABSTRACT

Bacterial infection is a great threat to human health. Lateral flow immunoassays (LFIAs) with the merits of low cost, quick screening, and on-site detection are competitive technologies for bacteria detection, but their detection limits depend on the optical performance of the adopted nanotags. Herein, we presented a LFIA platform for bacteria detection using polydopamine (PDA) functionalized Au nanoparticles (denoted as Au@PDA) as the nanotag. The introduction of PDA could provide enhanced light absorption of Au, as well as numerous functional groups for conjugation. Small recognition molecules i.e. vancomycin (Van) and p-mercaptophenylboronic acid (PMBA) were covalently anchored to Au@PDA, and selected as the specific probes towards Gram-positive (G+) and Gram-negative (G-) bacteria, respectively. Taken Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) as the representative targets of G+ and G- bacteria, two LFA strips were successfully constructed based on the immuno-sandwich principle. They could quantitatively detect S. aureus and E. coli both down to 102 cfu/mL, a very competitive detection limit in comparison with other colorimetric or luminescent probes-based LFIAs. Furthermore, the proposed two strips were applied for the quantitative, accurate, and rapid detection of S. aureus and E. coli in food and human urine samples with good analytical results obtained. In addition, they were integrated as a screening platform for quick evaluation of diverse antibacterial agents within 3 h, which is remarkably shortened compared with that of the two traditional methods i.e. bacterial culture and plate-counting.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Gold Colloid , Indoles , Metal Nanoparticles , Polymers , Staphylococcus aureus , Indoles/chemistry , Polymers/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Immunoassay/methods , Escherichia coli/isolation & purification , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/immunology , Gold Colloid/chemistry , Metal Nanoparticles/chemistry , Limit of Detection , Humans , Microbial Sensitivity Tests , Gold/chemistry , Vancomycin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL