Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 19(12): e1011877, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38127952

ABSTRACT

Shiga-toxin producing Escherichia coli (STEC) infections can cause from bloody diarrhea to Hemolytic Uremic Syndrome. The STEC intestinal infection triggers an inflammatory response that can facilitate the development of a systemic disease. We report here that neutrophils might contribute to this inflammatory response by secreting Interleukin 1 beta (IL-1ß). STEC stimulated neutrophils to release elevated levels of IL-1ß through a mechanism that involved the activation of caspase-1 driven by the NLRP3-inflammasome and neutrophil serine proteases (NSPs). Noteworthy, IL-1ß secretion was higher at lower multiplicities of infection. This secretory profile modulated by the bacteria:neutrophil ratio, was the consequence of a regulatory mechanism that reduced IL-1ß secretion the higher were the levels of activation of both caspase-1 and NSPs, and the production of NADPH oxidase-dependent reactive oxygen species. Finally, we also found that inhibition of NSPs significantly reduced STEC-triggered IL-1ß secretion without modulating the ability of neutrophils to kill the bacteria, suggesting NSPs might represent pharmacological targets to be evaluated to limit the STEC-induced intestinal inflammation.


Subject(s)
Escherichia coli Infections , Escherichia coli O157 , Hemolytic-Uremic Syndrome , Interleukin-1beta , Shiga-Toxigenic Escherichia coli , Humans , Caspases , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Neutrophils , Interleukin-1beta/metabolism
2.
Methods Mol Biol ; 2255: 97-117, 2021.
Article in English | MEDLINE | ID: mdl-34033098

ABSTRACT

Neutrophils release web like-structures known as neutrophil extracellular traps (NETs) that ensnare and kill microorganisms. These networks are constituted of a DNA scaffold with associated antimicrobial proteins, which are released to the extracellular space as an effective mechanism to fight against invading microorganisms. In parallel with this beneficial role to avoid microbial dissemination and wall off infections, accumulating evidence supports that under certain circumstances, NETs can exert deleterious effects in inflammatory, autoimmune, and thrombotic pathologies. Research on NET properties and their role in pathophysiological processes is a rapidly evolving and expanding field. Here, we describe a combination of methods to achieve a successful in vitro NET visualization, semiquantification, and isolation.


Subject(s)
Cell Separation/methods , DNA/analysis , Extracellular Traps/metabolism , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence/methods , Pancreatic Elastase/analysis , Peroxidase/metabolism , Humans , In Vitro Techniques
3.
Autophagy ; 17(9): 2629-2638, 2021 09.
Article in English | MEDLINE | ID: mdl-32954947

ABSTRACT

Neutrophils infected with Mycobacterium tuberculosis (Mtb) predominate in tuberculosis patients' lungs. Neutrophils phagocytose the pathogen, but the mechanism of pathogen elimination is controversial. Macroautophagy/autophagy, a crucial mechanism for several neutrophil functions, can be modulated by immunological mediators. The costimulatory molecule SLAMF1 can act as a microbial sensor in macrophages being also able to interact with autophagy-related proteins. Here, we demonstrate for the first time that human neutrophils express SLAMF1 upon Mtb-stimulation. Furthermore, SLAMF1 was found colocalizing with LC3B+ vesicles, and activation of SLAMF1 increased neutrophil autophagy induced by Mtb. Finally, tuberculosis patients' neutrophils displayed reduced levels of SLAMF1 and lower levels of autophagy against Mtb as compared to healthy controls. Altogether, these results indicate that SLAMF1 participates in neutrophil autophagy during active tuberculosis.Abbreviations: AFB: acid-fast bacilli; BafA1: bafilomycin A1; CLL: chronic lymphocytic leukemia; DPI: diphenyleneiodonium; EVs: extracellular vesicles; FBS: fetal bovine serum; HD: healthy donors; HR: high responder (tuberculosis patient); IFNG: interferon gamma; IL1B: interleukin 1 beta; IL17A: interleukin 17A; IL8: interleukin 8; LR: low responder (tuberculosis patient); mAb: monoclonal antibody; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK14/p38: mitogen-activated protein kinase 14; Mtb: Mycobacterium tuberculosis; Mtb-Ag: Mycobacterium tuberculosis, Strain H37Rv, whole cell lysate; NETs: neutrophils extracellular traps; PPD: purified protein derivative; ROS: reactive oxygen species; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; SLAMF1: signaling lymphocytic activation molecule family member 1; TB: tuberculosis; TLR: toll like receptor.


Subject(s)
Autophagy , Neutrophils , Signaling Lymphocytic Activation Molecule Family Member 1 , Tuberculosis , Humans , Macrophages/metabolism , Mycobacterium tuberculosis , Neutrophils/cytology , Neutrophils/microbiology , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Tuberculosis/microbiology
4.
Toxins (Basel) ; 12(1)2020 01 14.
Article in English | MEDLINE | ID: mdl-31947665

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) strains are food-borne pathogens that can cause different clinical conditions. Shiga toxin 2a and/or 2c (Stx2)-producing E. coli O157:H7 is the serotype most frequently associated with severe human disease. In this work we analyzed the hypothesis that host cells participate in Stx2 production, cell damage, and inflammation during EHEC infection. With this aim, macrophage-differentiated THP-1 cells and the intestinal epithelial cell line HCT-8 were incubated with E. coli O157:H7. A time course analysis of cellular and bacterial survival, Stx2 production, stx2 transcription, and cytokine secretion were analyzed in both human cell lines. We demonstrated that macrophages are able to internalize and kill EHEC. Simultaneously, Stx2 produced by internalized bacteria played a major role in macrophage death. In contrast, HCT-8 cells were completely resistant to EHEC infection. Besides, macrophages and HCT-8 infected cells produce IL-1ß and IL-8 inflammatory cytokines, respectively. At the same time, bacterial stx2-specific transcripts were detected only in macrophages after EHEC infection. The interplay between bacteria and host cells led to Stx production, triggering of inflammatory response and cell damage, all of which could contribute to a severe outcome after EHEC infections.


Subject(s)
Escherichia coli O157 , Host Microbial Interactions , Immunomodulation/physiology , Shiga Toxins/toxicity , Cell Line , Cytokines , Enterohemorrhagic Escherichia coli , Escherichia coli Infections , Humans , Inflammation , Macrophages
5.
Immunology ; 153(2): 225-237, 2018 02.
Article in English | MEDLINE | ID: mdl-28888033

ABSTRACT

γδ T cells are non-conventional, innate-like T cells, characterized by a restricted T-cell receptor repertoire. They participate in protective immunity responses against extracellular and intracellular pathogens, tumour surveillance, modulation of innate and adaptive immune responses, tissue healing, epithelial cell maintenance and regulation of physiological organ function. In this study, we investigated the role of neutrophils during the activation of human blood γδ T cells through CD3 molecules. We found that the up-regulation of CD69 expression, and the production of interferon-γ and tumour necrosis factor-α induced by anti-CD3 antibodies was potentiated by neutrophils. We found that inhibition of caspase-1 and neutralization of interleukin-18 did not affect neutrophil-mediated modulation. By contrast, the treatment with serine protease inhibitors prevented the potentiation of γδ T-cell activation induced by neutrophils. Moreover, the addition of elastase to γδ T-cell culture increased their stimulation, and the treatment of neutrophils with elastase inhibitor prevented the effect of neutrophils on γδ T-cell activation. Furthermore, we demonstrated that the effect of elastase on γδ T cells was mediated through the protease-activated receptor, PAR1, because the inhibition of this receptor with a specific antagonist, RWJ56110, abrogated the effect of neutrophils on γδ T-cell activation.


Subject(s)
Leukocyte Elastase/immunology , Lymphocyte Activation , Neutrophil Activation/immunology , Neutrophils/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD3 Complex/immunology , Humans , Interferon-gamma/immunology , Lectins, C-Type/immunology , Neutrophils/cytology , Receptor, PAR-1/immunology , T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/immunology
6.
J Innate Immun ; 8(4): 400-11, 2016.
Article in English | MEDLINE | ID: mdl-27230920

ABSTRACT

Hemolytic uremic syndrome (HUS), a vascular disease characterized by hemolytic anemia, thrombocytopenia, and acute renal failure, is caused by enterohemorrhagic Shiga toxin (Stx)-producing bacteria, which mainly affect children. Besides Stx, the inflammatory response mediated by neutrophils (PMN) is essential to HUS evolution. PMN can release neutrophil extracellular traps (NET) composed of DNA, histones, and other proteins. Since NET are involved in infectious and inflammatory diseases, the aim of this work was to investigate the contribution of NET to HUS. Plasma from HUS patients contained increased levels of circulating free-DNA and nucleosomes in comparison to plasma from healthy children. Neutrophils from HUS patients exhibited a greater capacity to undergo spontaneous NETosis. NET activated human glomerular endothelial cells, stimulating secretion of the proinflammatory cytokines IL-6 and IL-8. Stx induced PMN activation as judged by its ability to trigger reactive oxygen species production, increase CD11b and CD66b expression, and induce NETosis in PMN from healthy donors. During HUS, NET can contribute to the inflammatory response and thrombosis in the microvasculature and thus to renal failure. Intervention strategies to inhibit inflammatory mechanisms mediated by PMN, such as NETosis, could have a potential therapeutic impact towards amelioration of the severity of HUS.


Subject(s)
Bacterial Infections/immunology , Endothelial Cells/immunology , Extracellular Traps/immunology , Hemolytic-Uremic Syndrome/immunology , Kidney/pathology , Neutrophils/immunology , Shiga Toxin/immunology , Acute Kidney Injury , Anemia, Hemolytic , Apoptosis , Cells, Cultured , Child , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Neutrophil Activation , Neutrophils/microbiology , Reactive Oxygen Species/metabolism , Thrombocytopenia
7.
Invest Ophthalmol Vis Sci ; 55(9): 6116-26, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25190648

ABSTRACT

PURPOSE: To evaluate the role of nuclear factor-κB (NF-κB) activation in eye drop preservative toxicity and the effect of topical NF-κB inhibitors on preservative-facilitated allergic conjunctivitis. METHODS: Balb/c mice were instilled ovalbumin (OVA) combined with benzalkonium chloride (BAK) and/or NF-κB inhibitors in both eyes. After immunization, T-cell responses and antigen-induced ocular inflammation were evaluated. Nuclear factor-κB activation and associated inflammatory changes also were assessed in murine eyes and in an epithelial cell line after BAK exposure. RESULTS: Benzalkonium chloride promoted allergic inflammation and leukocyte infiltration of the conjunctiva. Topical NF-κB inhibitors blocked the disruptive effect of BAK on conjunctival immunological tolerance and ameliorated subsequent ocular allergic reactions. In line with these findings, BAK induced NF-κB activation and the secretion of IL-6 and granulocyte-monocyte colony-stimulating factor in an epithelial cell line and in the conjunctiva of instilled mice. In addition, BAK favored major histocompatibility complex (MHC) II expression in cultured epithelial cells in an NF-κB-dependent fashion after interaction with T cells. CONCLUSIONS: Benzalkonium chloride triggers conjunctival epithelial NF-κB activation, which seems to mediate some of its immune side effects, such as proinflammatory cytokine release and increased MHC II expression. Breakdown of conjunctival tolerance by BAK favors allergic inflammation, and this effect can be prevented in mice by topical NF-κB inhibitors. These results suggest a new pharmacological target for preservative toxicity and highlight the importance of conjunctival tolerance in ocular surface homeostasis.


Subject(s)
Benzalkonium Compounds/toxicity , Conjunctiva/immunology , Conjunctivitis, Allergic/prevention & control , I-kappa B Proteins/pharmacology , Immune Tolerance/drug effects , NF-kappa B/antagonists & inhibitors , Preservatives, Pharmaceutical/toxicity , Administration, Topical , Animals , Blotting, Western , Cell Line , Coculture Techniques , Conjunctiva/cytology , Conjunctivitis, Allergic/chemically induced , Conjunctivitis, Allergic/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Female , Flow Cytometry , Hypersensitivity, Delayed/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Microscopy, Confocal , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Ovalbumin/toxicity , T-Lymphocytes/immunology
8.
Semin Immunopathol ; 35(4): 423-37, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23370701

ABSTRACT

Neutrophils not only play a critical role as a first line of defense against bacteria and fungi infections but also contribute to tissue injury associated with autoimmune and inflammatory diseases. Neutrophils are rapidly and massively recruited from the circulation into injured tissues displaying an impressive arsenal of toxic weapons. Although effective in their ability to kill pathogens, these weapons were equally effective to induce tissue damage. Therefore, the inflammatory activity of neutrophils must be regulated with exquisite precision and timing, a task mainly achieved through a complex network of mechanisms, which regulate neutrophil survival. Neutrophils have the shortest lifespan among leukocytes and usually die via apoptosis although new forms of cell death have been characterized over the last few years. The lifespan of neutrophils can be dramatically modulated by a large variety of agents such as cytokines, pathogens, danger-associated molecular patterns as well as by pharmacological manipulation. Recent findings shed light about the complex mechanisms responsible for the regulation of neutrophil survival in different physiological, pathological, and pharmacological scenarios. Here, we provide an updated review on the current knowledge and new findings in this field and discuss novel strategies that could be used to drive the resolution of neutrophil-mediated inflammatory diseases.


Subject(s)
Apoptosis/physiology , Neutrophils/immunology , Neutrophils/metabolism , Caspases/metabolism , Cell Survival/physiology , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Death Domain/metabolism , Signal Transduction
9.
Cytokine ; 57(2): 258-68, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22154780

ABSTRACT

The development of acidic environments is a hallmark of inflammatory processes of different etiology. We have previously shown that transient exposure to acidic conditions, similar to those encountered in vivo, induces the activation of neutrophils and the phenotypic maturation of dendritic cells. We here report that extracellular acidosis (pH 6.5) selectively stimulates the production and the secretion of IL-1ß by human monocytes without affecting the production of TNF-α, IL-6 and the expression of CD40, CD80, CD86, and HLA-DR. Stimulation of IL-1ß production by pH 6.5-treated monocytes was shown to be dependent on caspase-1 activity, and it was also observed using peripheral blood mononuclear cells instead of isolated monocytes. Contrasting with the results in monocytes, we found that pH 6.5 did not stimulate any production of IL-1ß by macrophages. Changes in intracellular pH seem to be involved in the stimulation of IL-1ß production. In fact, monocytes cultured at pH 6.5 undergo a fall in the values of intracellular pH while the inhibitor of the Na+/H+ exchanger, 5-(N-ethyl-N-isopropyl)amiloride induced both, a decrease in the values of intracellular pH and the stimulation of IL-1ß production. Real time quantitative PCR assays indicated that monocytes cultured either at pH 6.5 or in the presence of 5-(N-ethyl-N-isopropyl)amiloride expressed higher levels of pro-IL-1ß mRNA suggesting that low values of intracellular pH enhance the production of IL-1ß, at least in part, by stimulating the synthesis of its precursor.


Subject(s)
Extracellular Space/metabolism , Interleukin-1beta/biosynthesis , Monocytes/metabolism , Calcium/metabolism , Caspase 1/metabolism , Cell Survival , Cytosol/metabolism , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intracellular Space/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/enzymology , Phenotype
10.
Crit Rev Immunol ; 24(5): 363-84, 2004.
Article in English | MEDLINE | ID: mdl-15663364

ABSTRACT

Dendritic cells (DCs) are the most efficient antigen-presenting cells. They are activated in the periphery by conserved pathogen molecules and by inflammatory mediators produced by a variety of cell types in response to danger signals. It is widely appreciated that inflammatory responses in peripheral tissues are usually associated with the development of acidic microenvironments. Surprisingly, there are relatively few studies directed to analyze the effect of extracellular acidosis on the immune response. We focus on the influence of extracellular acidosis on the function of immature DCs. The results presented here show that acidosis activates DCs. It increases the acquisition of extracellular antigens for MHC class I-restricted presentation and the ability of antigen-pulsed DCs to induce both specific CD8+ CTL and B-cell responses. These findings may have important implications to our understanding of the mechanisms through which DCs sense the presence of infection or inflammation in nonlymphoid tissues.


Subject(s)
Acidosis/metabolism , Dendritic Cells/metabolism , Animals , Antigen Presentation/immunology , Antigens/immunology , Cell Movement/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Inflammation/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Membrane Proteins/immunology , Neoplasms/metabolism , Pinocytosis/immunology , Receptors, Antigen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...