Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 14: 1334211, 2024.
Article in English | MEDLINE | ID: mdl-38817444

ABSTRACT

Parasites possess remarkable abilities to evade and manipulate the immune response of their hosts. Echinococcus granulosus is a parasitic tapeworm that causes cystic echinococcosis in animals and humans. The hydatid fluid released by the parasite is known to contain various immunomodulatory components that manipulate host´s defense mechanism. In this study, we focused on understanding the effect of hydatid fluid on dendritic cells and its impact on autophagy induction and subsequent T cell responses. Initially, we observed a marked downregulation of two C-type lectin receptors in the cell membrane, CLEC9A and CD205 and an increase in lysosomal activity, suggesting an active cellular response to hydatid fluid. Subsequently, we visualized ultrastructural changes in stimulated dendritic cells, revealing the presence of macroautophagy, characterized by the formation of autophagosomes, phagophores, and phagolysosomes in the cell cytoplasm. To further elucidate the underlying molecular mechanisms involved in hydatid fluid-induced autophagy, we analyzed the expression of autophagy-related genes in stimulated dendritic cells. Our results demonstrated a significant upregulation of beclin-1, atg16l1 and atg12, indicating the induction of autophagy machinery in response to hydatid fluid exposure. Additionally, using confocal microscopy, we observed an accumulation of LC3 in dendritic cell autophagosomes, confirming the activation of this catabolic pathway associated with antigen presentation. Finally, to evaluate the functional consequences of hydatid fluid-induced autophagy in DCs, we evaluated cytokine transcription in the splenocytes. Remarkably, a robust polyfunctional T cell response, with inhibition of Th2 profile, is characterized by an increase in the expression of il-6, il-10, il-12, tnf-α, ifn-γ and tgf-ß genes. These findings suggest that hydatid fluid-induced autophagy in dendritic cells plays a crucial role in shaping the subsequent T cell responses, which is important for a better understanding of host-parasite interactions in cystic echinococcosis.


Subject(s)
Autophagy , Dendritic Cells , Echinococcosis , Echinococcus granulosus , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Echinococcus granulosus/immunology , Autophagy/immunology , Echinococcosis/immunology , Echinococcosis/parasitology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice , Lectins, C-Type/metabolism , Cytokines/metabolism , Female , Autophagosomes/immunology , Autophagosomes/metabolism
2.
Parasit Vectors ; 16(1): 255, 2023 Jul 29.
Article in English | MEDLINE | ID: mdl-37516852

ABSTRACT

BACKGROUND: The Echinococcus granulosus sensu lato species complex causes cystic echinococcosis, a zoonotic disease of medical importance. Parasite-derived small extracellular vesicles (sEVs) are involved in the interaction with hosts intervening in signal transduction related to parasite proliferation and disease pathogenesis. Although the characteristics of sEVs from E. granulosus protoscoleces and their interaction with host dendritic cells (DCs) have been described, the effect of sEVs recovered during parasite pharmacological treatment on the immune response remains unexplored. METHODS: Here, we isolated and characterized sEVs from control and drug-treated protoscoleces by ultracentrifugation, transmission electron microscopy, dynamic light scattering, and proteomic analysis. In addition, we evaluated the cytokine response profile induced in murine bone marrow-derived dendritic cells (BMDCs) by qPCR. RESULTS: The isolated sEVs, with conventional size between 50 and 200 nm, regardless of drug treatment, showed more than 500 cargo proteins and, importantly, 20 known antigens and 70 potential antigenic proteins, and several integral-transmembrane and soluble proteins mainly associated with signal transduction, immunomodulation, scaffolding factors, extracellular matrix-anchoring, and lipid transport. The identity and abundance of proteins in the sEV-cargo from metformin- and albendazole sulfoxide (ABZSO)-treated parasites were determined by proteomic analysis, detecting 107 and eight exclusive proteins, respectively, which include proteins related to the mechanisms of drug action. We also determined that the interaction of murine BMDCs with sEVs derived from control parasites and those treated with ABZSO and metformin increased the expression of pro-inflammatory cytokines such as IL-12 compared to control cells. Additionally, protoscolex-derived vesicles from metformin treatments induced the production of IL-6, TNF-α, and IL-10. However, the expression of IL-23 and TGF-ß was downregulated. CONCLUSIONS: We demonstrated that sEV-cargo derived from drug-treated E. granulosus protoscoleces have immunomodulatory functions, as they enhance DC activation towards a type 1 pro-inflammatory profile against the parasite, and therefore support the proposal of a new approach for the prevention and treatment of secondary echinococcosis.


Subject(s)
Echinococcosis , Echinococcus granulosus , Echinococcus , Extracellular Vesicles , Animals , Mice , Proteomics , Signal Transduction , Echinococcosis/drug therapy , Immunity
3.
Antimicrob Agents Chemother ; 67(1): e0114022, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36533935

ABSTRACT

Osteomyelitis caused by Staphylococcus aureus is an important and current health care problem worldwide. Treatment of this infection frequently fails not only due to the increasing incidence of antimicrobial-resistant isolates but also because of the ability of S. aureus to evade the immune system, adapt to the bone microenvironment, and persist within this tissue for decades. We have previously demonstrated the role of staphylococcal protein A (SpA) in the induction of exacerbated osteoclastogenesis and increased bone matrix degradation during osteomyelitis. The aim of this study was to evaluate the potential of using anti-SpA antibodies as an adjunctive therapy to control inflammation and bone damage. By using an experimental in vivo model of osteomyelitis, we demonstrated that the administration of an anti-SpA antibody by the intraperitoneal route prevented excessive inflammatory responses in the bone upon challenge with S. aureus. Ex vivo assays indicated that blocking SpA reduced the priming of osteoclast precursors and their response to RANKL. Moreover, the neutralization of SpA was able to prevent the differentiation and activation of osteoclasts in vivo, leading to reduced expression levels of cathepsin K, reduced expression of markers associated with abnormal bone formation, and decreased trabecular bone loss during osteomyelitis. Taken together, these results demonstrate the feasibility of using anti-SpA antibodies as an antivirulence adjunctive therapy that may prevent the development of pathological conditions that not only damage the bone but also favor bacterial escape from antimicrobials and the immune system.


Subject(s)
Osteomyelitis , Staphylococcal Infections , Humans , Osteoclasts/metabolism , Osteoclasts/pathology , Staphylococcus aureus , Staphylococcal Protein A/metabolism , Osteomyelitis/drug therapy , Osteomyelitis/microbiology , Osteogenesis , Staphylococcal Infections/microbiology
4.
Front Immunol ; 11: 524180, 2020.
Article in English | MEDLINE | ID: mdl-33692774

ABSTRACT

The type 1 TNF-α receptor (TNFR1) has a central role in initiating both pro-inflammatory and pro-apoptotic signaling cascades in neutrophils. Considering that TNFR1 signals Staphylococcus aureus protein A (SpA), the aim of this study was to explore the interaction of this bacterial surface protein with neutrophils and keratinocytes to underscore the signaling pathways that may determine the fate of these innate immune cells in the infected tissue during staphylococcal skin infections. Using human neutrophils cultured in vitro and isogenic staphylococcal strains expressing or not protein A, we demonstrated that SpA is a potent inducer of IL-8 in neutrophils and that the induction of this chemokine is dependent on the SpA-TNFR1 interaction and p38 activation. In addition to IL-8, protein A induced the expression of TNF-α and MIP-1α highlighting the importance of SpA in the amplification of the inflammatory response. Protein A contributed to reduce neutrophil mortality prolonging their lifespan upon the encounter with S. aureus. Signaling initiated by SpA modulated the type of neutrophil cell death in vitro and during skin and soft tissue infections (SSTI) in vivo triggering the apoptotic pathway instead of necrosis. Moreover, SpA induced pro-inflammatory cytokines in keratinocytes, modulating their survival in vitro and preventing the exacerbated necrosis and ulceration of the epithelium during SSTI in vivo. Taken together, these results highlight the importance of the inflammatory signaling induced by protein A in neutrophils and skin epithelial cells. The ability of protein A to modulate the neutrophil/epithelial cell death program in the skin is of clinical relevance considering that lysis of neutrophils and epithelial cells will promote an intense inflammatory response and contribute to tissue damage, a non-desirable feature of complicated SSTI.


Subject(s)
Keratinocytes/immunology , MAP Kinase Signaling System/immunology , Neutrophils/immunology , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology , Cytokines/immunology , Humans , Keratinocytes/microbiology , Neutrophils/microbiology , Receptors, Tumor Necrosis Factor, Type I/immunology , p38 Mitogen-Activated Protein Kinases/immunology
5.
Front Immunol ; 10: 2374, 2019.
Article in English | MEDLINE | ID: mdl-31681277

ABSTRACT

Fever is a hallmark of infections and inflammatory diseases, represented by an increase of 1-4°C in core body temperature. Fever-range hyperthermia (FRH) has been shown to increase neutrophil recruitment to local sites of infection. Here, we evaluated the impact of a short period (1 h) of FRH (STFRH) on pro-inflammatory and bactericidal human neutrophil functions. STFRH did not affect neutrophil spontaneous apoptosis but reverted the lipopolysaccharide (LPS)-induced anti-apoptotic effect compared with that under normothermic conditions. Furthermore, STFRH accelerated phorbol myristate acetate (PMA)-induced NETosis evaluated either by the nuclear DNA decondensation at 2 h post-stimulation or by the increase in extracellular DNA that colocalized with myeloperoxidase (MPO) at 4 h post-stimulation. Increased NETosis upon STFRH was associated with an increase in reactive oxygen species (ROS) production but not in autophagy levels. STFRH also increased NETosis in response to Pseudomonas aeruginosa challenge but moderately reduced its phagocytosis. However, these STFRH-induced effects did not influence the ability of neutrophils to kill bacteria after 4 h of co-culture. STFRH also significantly reduced neutrophil capacity to release the pro-inflammatory cytokines chemokine (C-X-C motif) ligand 8/interleukin 8 (CXCL8/IL-8) and IL-1ß in response to LPS and P. aeruginosa challenge. Altogether, these results indicate that a short and mild hyperthermal period is enough to modulate neutrophil responses to bacterial encounter. They also suggest that fever spikes during bacterial infections might lead neutrophils to trigger an emergency response promoting neutrophil extracellular trap (NET) formation to ensnare bacteria in order to wall off the infection and to reduce their release of pro-inflammatory cytokines in order to limit the inflammatory response.


Subject(s)
Extracellular Traps/immunology , Fever/immunology , Interleukin-1beta/immunology , Interleukin-8/immunology , Neutrophils/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Extracellular Traps/microbiology , Female , Fever/microbiology , Fever/pathology , Humans , Male , Neutrophils/microbiology , Neutrophils/pathology , Pseudomonas Infections/pathology
6.
Biochim Biophys Acta Mol Basis Dis ; 1865(10): 2657-2670, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31299217

ABSTRACT

Staphylococcus aureus is the most prominent cause of skin and soft tissue infections (SSTI) worldwide. Mortality associated with invasive SSTI is a major threat to public health considering the incidence of antibiotic resistant isolates in particular methicillin resistant S. aureus both in the hospital (HA-MRSA) and in the community (CA-MRSA). To overcome the increasing difficulties in the clinical management of SSTI due to MRSA, new prophylactic and therapeutic approaches are urgently needed and a preventive vaccine would be welcome. The rational design of an anti-S. aureus vaccine requires a deep knowledge of the role that the different bacterial virulence factors play according to the type of infection. In the present study, using a set of isogenic deficient mutants and their complemented strains we determined that the staphylococcal surface proteins SpA and Sbi play an important role in the induction of inflammatory cytokines and chemokines in the skin during SSTI. SpA and Sbi initiate signaling cascades that lead to the early recruitment of neutrophils, modulate their lifespan in the skin milieu and contribute to proper abscess formation and bacterial eradication. Moreover, the expression of SpA and Sbi appear critical for skin repair and wound healing. Thus, these results indicate that SpA and Sbi can promote immune responses in the skin that are beneficial for the host and therefore, should not be neutralized with vaccine formulations designed to prevent SSTI.


Subject(s)
Abscess/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Skin/immunology , Soft Tissue Infections/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Wound Healing/physiology , Abscess/metabolism , Abscess/microbiology , Animals , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Humans , Keratinocytes , Methicillin-Resistant Staphylococcus aureus/metabolism , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin/microbiology , Skin/pathology , Soft Tissue Infections/immunology , Soft Tissue Infections/microbiology , Soft Tissue Infections/pathology , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity
7.
Article in English | MEDLINE | ID: mdl-30123776

ABSTRACT

Early research on sepsis has focused on the initial hyper-inflammatory, cytokine mediated phase of the disorder whereas the events that govern the concomitant and subsequent anti-inflammatory compensatory response are not completely understood. In this context, the putative participation of TNFR1-mediated signaling in the immunosuppressive phase of Staphylococcus aureus sepsis has not been elucidated. The aim of this study was to determine the role of TNFR1 in directing the immune dysfunction during S. aureus sepsis and the potential contribution of MDSC to this process. Using a model of sepsis of peritoneal origin and tnfr1-/- mice, we demonstrated that during staphylococcal sepsis CD4+ T cell anergy is significantly dependent on TNFR1 expression and that signaling through this receptor has an impact on bacterial clearance in the spleen. MDSC played a major role in the generation of anergic CD4+ T cells and their accumulation in the spleen during S. aureus sepsis correlated with IL-6 induction. Although TNFR1 signaling was not required for MDSC accumulation and expansion in the spleen, it determined the in vivo expression of Arginase 1 and iNOS, enzymes known to participate in the suppressive function of this population. Moreover, our data indicate that TNFR1-mediated IL-10 production may modulate MDSC function during staphylococcal sepsis. Taken together these results indicate that TNFR1 plays a critical role on T cell dysfunction during S. aureus sepsis by regulating immunomodulatory mediators in MDSC. The role of TNFR1-mediated signaling during the immunosuppressive phase of staphylococcal sepsis should be considered when designing novel alternative therapeutic approaches.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunosuppression Therapy , Myeloid-Derived Suppressor Cells/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sepsis/pathology , Signal Transduction , Staphylococcal Infections/pathology , Animals , Arginase/metabolism , Interleukin-6/metabolism , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Sepsis/immunology , Spleen/pathology , Staphylococcal Infections/immunology
8.
Immunology ; 154(3): 510-521, 2018 07.
Article in English | MEDLINE | ID: mdl-29377107

ABSTRACT

Ultraviolet radiation (UVr) promotes several well-known molecular changes, which may ultimately impact on health. Some of these effects are detrimental, like inflammation, carcinogenesis and immunosuppression. On the other hand, UVr also promotes vitamin D synthesis and other beneficial effects. We recently demonstrated that exposure to very low doses of UVr on four consecutive days [repetitive low UVd (rlUVd)] does not promote an inflammatory state, nor the recruitment of neutrophils or lymphocytes, as the exposure to a single high UV dose (shUVd) does. Moreover, rlUVd reinforce the epithelium by increasing antimicrobial peptides transcription and epidermal thickness. The aim of this study was to evaluate the adaptive immune response after shUVd and rlUVd, determining T-cell and B-cell responses. Finally, we challenged animals exposed to both irradiation procedures with Staphylococcus aureus to study the overall effects of both innate and adaptive immunity during a cutaneous infection. We observed, as expected, a marked suppression of T-cell and B-cell responses after exposure to an shUVd but a novel and significant increase in both specific responses after exposure to rlUVd. However, the control of the cutaneous S. aureus infection was defective in this last group, suggesting that responses against pathogens cannot be ruled out from isolated stimuli.


Subject(s)
Adaptive Immunity/radiation effects , Radiation Exposure , Ultraviolet Rays , Animals , Antibody Formation/immunology , Antibody Formation/radiation effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Biomarkers , Cytokines/metabolism , Dermatitis/immunology , Dermatitis/metabolism , Dermatitis/microbiology , Dermatitis/prevention & control , Disease Models, Animal , Immunization , Immunophenotyping , Male , Mice , Radiation Dosage , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/prevention & control , Staphylococcus aureus/immunology , Staphylococcus aureus/radiation effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/immunology
9.
Int J Med Microbiol ; 307(4-5): 191-199, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28549830

ABSTRACT

Community-acquired methicillin resistant Staphylococcus aureus emerged as a worldwide health problem in the last few years. In Argentina, it is found in 70% of skin and skin structure infections in previously healthy adult patients and causes severe invasive diseases. The ST30-SCCmecIVc-spat019 clone is predominant in adult infections and has displaced the previously prevalent ST5-SCCmecIVa-spat311 clone in community settings. In the present work we compared the virulence of both clones in order to explain the displacement, and found that ST30-IVc is associated with invasive infections in adult patients from Argentina and possesses a different virulence-associated genes profile compared to ST5-IVa. A representative strain of ST30 lineage has a more aggressive behavior in animal models of infection and expresses higher level of Fibronectin binding protein A coding gene, which could enhance the bacterial invasion capacity.


Subject(s)
Bacterial Proteins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Virulence Factors/genetics , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Argentina , Bacterial Proteins/metabolism , Colony Count, Microbial , Cross Infection/microbiology , Disease Models, Animal , Female , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Multicenter Studies as Topic , Rats , Rats, Wistar , Respiratory Tract Infections/microbiology , Staphylococcal Skin Infections/microbiology
10.
PLoS One ; 10(6): e0131879, 2015.
Article in English | MEDLINE | ID: mdl-26126119

ABSTRACT

Staphylococcus aureus is an important human pathogen that causes infections that may present high morbidity and mortality. Among its many virulence factors protein A (SpA) and Staphylococcal binding immunoglobulin protein (Sbi) bind the Fc portion of IgG interfering with opsonophagocytosis. We have previously demonstrated that SpA interacts with the TNF-α receptor (TNFR) 1 through each of the five IgG binding domains and induces the production of pro-inflammatory cytokines and chemokines. The IgG binding domains of Sbi are homologous to those of SpA, which allow us to hypothesize that Sbi might also have a role in the inflammatory response induced by S. aureus. We demonstrate that Sbi is a novel factor that participates in the induction of the inflammatory response during staphylococcal infections via TNFR1 and EGFR mediated signaling as well as downstream MAPKs. The expression of Sbi significantly contributed to IL-6 production and modulated CXCL-1 expression as well as neutrophil recruitment to the site of infection, thus demonstrating for the first time its relevance as a pro-inflammatory staphylococcal antigen in an in vivo model.


Subject(s)
Bacterial Proteins/immunology , Carrier Proteins/immunology , Inflammation/immunology , Staphylococcal Infections/immunology , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology , Animals , Binding Sites, Antibody/immunology , Chemokine CXCL1/biosynthesis , ErbB Receptors/immunology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Flavonoids/pharmacology , Imidazoles/pharmacology , Immunoglobulin G/immunology , Inflammation/microbiology , Interleukin-6/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neutrophil Infiltration/immunology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction/immunology , Tyrphostins/pharmacology , Virulence Factors/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
11.
Infect Immun ; 81(11): 4200-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002060

ABSTRACT

Staphylococcus aureus infections are an important public health concern due to their increasing incidence and high rates of mortality. The success of S. aureus as a pathogen is highly related to its enormous capacity to evade the host immune response. The critical role of tumor necrosis factor alpha (TNF-α) in the initial host defense against systemic staphylococcal infection has been demonstrated in experimental models and may partially explain the lack of significant benefits observed in clinical trials attempting to neutralize this cytokine in septic patients. S. aureus protein A plays a key role in regulating inflammation through its ability to bind and signal through the TNF-α receptor 1 (TNFR1). In this study, we demonstrate that S. aureus, via protein A-mediated signaling, induces early shedding of TNFR1, which precedes the secretion of TNF-α in vitro and in vivo. The results obtained using a protein A-deficient mutant and tnfr1(-/-) mice strongly suggest that the increased levels of soluble TNFR1 present during experimental S. aureus infection may neutralize circulating TNF-α and impair the host inflammatory response. Early shedding of TNFR1 induced by protein A may constitute a novel mechanism by which S. aureus subverts the host immune response.


Subject(s)
Receptors, Tumor Necrosis Factor, Type I/metabolism , Sepsis/immunology , Staphylococcal Infections/immunology , Staphylococcal Protein A/metabolism , Staphylococcus aureus/immunology , Tumor Necrosis Factor-alpha/immunology , Virulence Factors/metabolism , Animals , Cell Line , Humans , Immune Evasion , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Sepsis/microbiology , Staphylococcus aureus/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...