Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(10): 1503-1513, 2019 10.
Article in English | MEDLINE | ID: mdl-31163264

ABSTRACT

Outer membrane vesicles (OMVs) are secreted by Gram-negative bacteria and induce a stronger inflammatory response than pure LPS. After endocytosis of OMVs by macrophages, lipopolysaccharide (LPS) is released from early endosomes to activate its intracellular receptors followed by non-canonical inflammasome activation and pyroptosis, which are critically involved in sepsis development. Previously, we could show that the synthetic anti-endotoxin peptide Pep19-2.5 neutralizes inflammatory responses induced by intracellular LPS. Here, we aimed to investigate whether Pep19-2.5 is able to suppress cytoplasmic LPS-induced inflammation under more physiological conditions by using OMVs which naturally transfer LPS to the cytosol. Isothermal titration calorimetry revealed an exothermic reaction between Pep19-2.5 and Escherichia coli OMVs and the Limulus Amebocyte Lysate assay indicated a strong endotoxin blocking activity. In THP-1 macrophages and primary human macrophages Pep19-2.5 and polymyxin B reduced interleukin (IL)-1ß and tumor necrosis factor (TNF) release as well as pyroptosis induced by OMVs, while the Toll-like receptor 4 signaling inhibitor TAK-242 suppressed OMV-induced TNF and IL-1ß secretion, but not pyroptosis. Internalization of Pep19-2.5 was at least partially mediated by the P2X7 receptor in macrophages but not in monocytes. Additionally, a cell-dependent difference in the neutralization efficiency of Pep19-2.5 became evident in macrophages and monocytes, indicating a critical role for peptide-mediated IL-1ß secretion via the P2X7 receptor. In conclusion, we provide evidence that LPS-neutralizing peptides inhibit OMV-induced activation of the inflammasome/IL-1 axis and give new insights into the mechanism of peptide-mediated neutralization of cytoplasmic LPS suggesting an essential and cell-type specific role for the P2X7 receptor.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bacterial Outer Membrane/drug effects , Escherichia coli/drug effects , Lipopolysaccharides/antagonists & inhibitors , Macrophages/drug effects , Peptides/pharmacology , Bacterial Outer Membrane/immunology , Cell Line , Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/microbiology , Lipopolysaccharides/immunology , Macrophages/immunology , Pyroptosis/drug effects
2.
Trends Pharmacol Sci ; 40(3): 187-197, 2019 03.
Article in English | MEDLINE | ID: mdl-30691865

ABSTRACT

Lipopolysaccharide (LPS) sensing in the cytosol by the noncanonical inflammasome leads to pyroptosis and NLRP3 inflammasome activation. This mechanism may be more critical for sepsis development than recognition of LPS by Toll-like receptor 4. LPS is directly binding to its intracellular receptor caspase-4/5/11, mediated by outer membrane vesicles and guanylate-binding proteins that deliver LPS to the cytosol and mediate access of caspases to LPS. Caspase-11-dependent cleavage of gasdermin D is discussed as a link between LPS-induced activation of caspases and pyroptosis or NLRP3 inflammasome activation. Finally, we highlight recently described inhibitors of cytosolic LPS-triggered noncanonical inflammasome activation that might be considered as potential drugs for the treatment of sepsis.


Subject(s)
Lipopolysaccharides/metabolism , Sepsis/metabolism , Animals , Caspases/metabolism , Humans , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins , Molecular Targeted Therapy , Neoplasm Proteins/metabolism , Phosphate-Binding Proteins , Sepsis/drug therapy
3.
Br J Pharmacol ; 175(17): 3581-3593, 2018 09.
Article in English | MEDLINE | ID: mdl-29947028

ABSTRACT

BACKGROUND AND PURPOSE: Wound healing is a complex process that is essential to provide skin homeostasis. Infection with pathogenic bacteria such as Staphylococcus aureus can lead to chronic wounds, which are challenging to heal. Previously, we demonstrated that the antimicrobial endotoxin-neutralizing peptide Pep19-2.5 promotes artificial wound closure in keratinocytes. Here, we investigated the mechanism of peptide-induced cell migration and if Pep19-2.5 accelerates wound closure in vivo. EXPERIMENTAL APPROACH: Cell migration was examined in HaCaT keratinocytes and P2X7 receptor-overexpressing HEK293 cells using the wound healing scratch assay. The protein expression of phosphorylated ERK1/2, ATP release, calcium influx and mitochondrial ROS were analysed to characterize Pep19-2.5-mediated signalling. For in vivo studies, female BALB/c mice were wounded and infected with methicillin-resistant S. aureus (MRSA) or left non-infected and treated topically with Pep19-2.5 twice daily for 6 days. KEY RESULTS: Specific P2X7 receptor antagonists inhibited Pep19-2.5-induced cell migration and ERK1/2 phosphorylation in keratinocytes and P2X7 receptor-transfected HEK293 cells. ATP release was not increased by Pep19-2.5; however, ATP was required for cell migration. Pep19-2.5 increased cytosolic calcium and mitochondrial ROS, which were involved in peptide-induced migration and ERK1/2 phosphorylation. In both non-infected and MRSA-infected wounds, the wound diameter was reduced already at day 2 post-wounding in the Pep19-2.5-treated groups compared to vehicle, and remained decreased until day 6. CONCLUSIONS AND IMPLICATIONS: Our data suggest the potential application of Pep19-2.5 in the treatment of non-infected and S. aureus-infected wounds and provide insights into the mechanism involved in Pep19-2.5-induced wound healing.


Subject(s)
Anti-Bacterial Agents/pharmacology , Endotoxins/antagonists & inhibitors , Keratinocytes/drug effects , Peptides/pharmacology , Purinergic Agonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/chemistry , Cell Line, Transformed , Female , HEK293 Cells , Humans , Keratinocytes/cytology , Mice , Mice, Inbred BALB C
4.
Front Pharmacol ; 9: 281, 2018.
Article in English | MEDLINE | ID: mdl-29643807

ABSTRACT

Alarming data about increasing resistance to conventional antibiotics are reported, while at the same time the development of new antibiotics is stagnating. Skin and soft tissue infections (SSTIs) are mainly caused by the so called ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) which belong to the most recalcitrant bacteria and are resistant to almost all common antibiotics. S. aureus and P. aeruginosa are the most frequent pathogens isolated from chronic wounds and increasing resistance to topical antibiotics has become a major issue. Therefore, new treatment options are urgently needed. In recent years, research focused on the development of synthetic antimicrobial peptides (AMPs) with lower toxicity and improved activity compared to their endogenous counterparts. AMPs appear to be promising therapeutic options for the treatment of SSTIs and wounds as they show a broad spectrum of antimicrobial activity, low resistance rates and display pivotal immunomodulatory as well as wound healing promoting activities such as induction of cell migration and proliferation and angiogenesis. In this review, we evaluate the potential of AMPs for the treatment of bacterial SSTIs and wounds and provide an overview of the mechanisms of actions of AMPs that contribute to combat skin infections and to improve wound healing. Bacteria growing in biofilms are more resistant to conventional antibiotics than their planktonic counterparts due to limited biofilm penetration and distinct metabolic and physiological functions, and often result in chronification of infections and wounds. Thus, we further discuss the feasibility of AMPs as anti-biofilm agents. Finally, we highlight perspectives for future therapies and which issues remain to bring AMPs successfully to the market.

5.
Biochem Pharmacol ; 140: 64-72, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28539262

ABSTRACT

Toll-like receptor (TLR) 4-independent recognition of lipopolysaccharide (LPS) in the cytosol by inflammatory caspases leads to non-canonical inflammasome activation and induction of IL-1 secretion and pyroptosis. The discovery of this novel mechanism has potential implications for the development of effective drugs to treat sepsis since LPS-mediated hyperactivation of caspases is critically involved in endotoxic shock. Previously, we demonstrated that Pep19-2.5, a synthetic anti-endotoxin peptide, efficiently neutralises pathogenicity factors of Gram-negative and Gram-positive bacteria and protects against sepsis in vivo. Here, we report that Pep19-2.5 inhibits the effects of cytoplasmic LPS in human myeloid cells and keratinocytes. In THP-1 monocytes and macrophages, the peptide strongly reduced secretion of IL-1ß and LDH induced by intracellular LPS. In contrast, the TLR4 signaling inhibitor TAK-242 abrogates LPS-induced TNF and IL-1ß secretion, but not pyroptotic cell death. Furthermore, Pep19-2.5 suppressed LPS-induced HMGB-1 production and caspase-1 activation in THP-1 monocytes. Consistent with this observation, we found impaired IL-1ß and IL-1α release in LPS-stimulated primary monocytes in the presence of Pep19-2.5 and reduced LDH release and IL-1B and IL-1A expression in LPS-transfected HaCaT keratinocytes. Additionally, Pep19-2.5 completely abolished IL-1ß release induced by LPS/ATP in macrophages via canonical inflammasome activation. In conclusion, we provide evidence that anti-endotoxin peptides inhibit the inflammasome/IL-1 axis induced by cytoplasmic LPS sensing in myeloid cells and keratinocytes and activation of the classical inflammasome by LPS/ATP which may contribute to the protection against bacterial sepsis and skin infections with intracellular Gram-negative bacteria.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammasomes/drug effects , Keratinocytes/drug effects , Macrophages/drug effects , Peptides/pharmacology , Pyroptosis/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cytoplasm/drug effects , Cytoplasm/metabolism , Endotoxins/antagonists & inhibitors , Endotoxins/toxicity , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Peptides/chemistry , Sulfonamides/pharmacology , Toll-Like Receptor 4/metabolism
6.
J Steroid Biochem Mol Biol ; 171: 201-208, 2017 07.
Article in English | MEDLINE | ID: mdl-28377308

ABSTRACT

Glucocorticoids (GCs) induce Toll-like receptor (TLR) 2 expression and synergistically upregulate TLR2 with pro-inflammatory cytokines or bacteria. These paradoxical effects have drawn attention to the inflammatory initiating or promoting effects of GCs, as GC treatment can provoke inflammatory skin diseases. Here, we aimed to investigate the regulatory effects of GCs in human skin cells of different epidermal and dermal layers. We found that Dex induced TLR2 expression mainly in undifferentiated and less in calcium-induced differentiated keratinocytes but not in HaCaT cells or fibroblasts, however, Dex reduced TLR1/6 expression. Stimulation with Dex under inflammatory conditions further increased TLR2 but not TLR1 or TLR6 levels in keratinocytes. Increased ligand-induced interaction of TLR2 with MyD88 and expression of the adaptor protein TRAF6 indicated enhanced TLR2 signalling, whereas TLR2/1 or TLR2/6 signalling was not increased in Dex-pretreated keratinocytes. GC-increased TLR2 expression was negatively regulated by JNK MAPK signalling when stimulated with Propionibacterium acnes. Our results provide novel insights into the molecular mechanisms of glucocorticoid-mediated expression and function of TLR2 in human skin cells and the understanding of the mechanisms of corticosteroid side effects.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Dexamethasone/adverse effects , Gene Expression Regulation/drug effects , Glucocorticoids/adverse effects , MAP Kinase Signaling System/drug effects , Skin/drug effects , Toll-Like Receptor 2/agonists , Cell Line , Cells, Cultured , Child , Dermis/cytology , Dermis/drug effects , Dermis/immunology , Dermis/metabolism , Epidermal Cells , Epidermis/drug effects , Epidermis/immunology , Epidermis/metabolism , Humans , Interleukin-8/agonists , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Intracellular Signaling Peptides and Proteins , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Myeloid Differentiation Factor 88/metabolism , Propionibacterium acnes/immunology , Skin/cytology , Skin/immunology , Skin/metabolism , TNF Receptor-Associated Factor 6/agonists , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 1/antagonists & inhibitors , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/antagonists & inhibitors , Toll-Like Receptor 6/genetics , Toll-Like Receptor 6/metabolism
7.
Sci Rep ; 6: 31577, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27509895

ABSTRACT

The stagnation in the development of new antibiotics and the concomitant high increase of resistant bacteria emphasize the urgent need for new therapeutic options. Antimicrobial peptides are promising agents for the treatment of bacterial infections and recent studies indicate that Pep19-2.5, a synthetic anti-lipopolysaccharide (LPS) peptide (SALP), efficiently neutralises pathogenicity factors of Gram-negative (LPS) and Gram-positive (lipoprotein/-peptide, LP) bacteria and protects against sepsis. Here, we investigated the potential of Pep19-2.5 and the structurally related compound Pep19-4LF for their therapeutic application in bacterial skin infections. SALPs inhibited LP-induced phosphorylation of NF-κB p65 and p38 MAPK and reduced cytokine release and gene expression in primary human keratinocytes and dermal fibroblasts. In LPS-stimulated human monocyte-derived dendritic cells and Langerhans-like cells, the peptides blocked IL-6 secretion, downregulated expression of maturation markers and inhibited dendritic cell migration. Both SALPs showed a low cytotoxicity in all investigated cell types. Furthermore, SALPs markedly promoted cell migration via EGFR transactivation and ERK1/2 phosphorylation and accelerated artificial wound closure in keratinocytes. Peptide-induced keratinocyte migration was mediated by purinergic receptors and metalloproteases. In contrast, SALPs did not affect proliferation of keratinocytes. Conclusively, our data suggest a novel therapeutic target for the treatment of patients with acute and chronic skin infections.


Subject(s)
Cell Movement/drug effects , Cell Movement/physiology , Immunologic Factors/metabolism , Keratinocytes/drug effects , Keratinocytes/physiology , Peptides/metabolism , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/physiology , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Lipopolysaccharides/antagonists & inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Dermatol Sci ; 82(1): 9-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26803226

ABSTRACT

BACKGROUND: As mediators between innate and adaptive immune responses, Langerhans cells (LCs) are in the focus of recent investigations to determine their role in allergic inflammatory diseases like allergic contact dermatitis and atopic dermatitis. Sphingosine 1-phosphate (S1P) is a crucial lipid mediator in the skin and potentially interferes with LC homeostasis but also functional properties, such as cytokine release, migration and antigen-uptake which are considered to be key events in the initiation and maintenance of pathological disorders. OBJECTIVE: Here, we used human Langerhans-like cells to study the influence of S1P-mediated signalling on LC maturation, cytokine release, migration and endocytosis. METHODS: Immature Langerhans-like cells were generated from the human acute myeloid leukaemia cell line MUTZ-3 (MUTZ-LCs) and human primary monocytes (MoLCs). S1P receptor expression was determined by quantitative RT-PCR and western blotting. Expression of maturation markers were investigated by flow cytometry. The influence of S1P signalling on cytokine release was quantified by ELISA. Migration assays and FITC-dextran uptake in the presence of S1P, specific S1 P receptor agonists and antagonists as well as fingolimod (FTY720) were analysed through fluorescence microscopy and flow cytometry. RESULTS: S1P receptor protein expression was confirmed for S1P1, S1P2 and S1P4 in MUTZ-LCs and S1P1 and S1P2 in MoLCs. In mature cells S1P receptors were downregulated. S1P did not induce maturation in MUTZ-LCs, whereas in MoLCs CD83 and CD86 were slightly upregulated. IL-8 release of MUTZ-LCs matured in the presence of S1P was not altered, however, reduced IL-6 and IL-12p70 levels were observed in mature MoLCs. Interestingly, immature MUTZ-LCs revealed a significantly increased S1P-dependent migratory capacity, whereas CCL20 induced migration was significantly decreased in the presence of S1P. Furthermore, migratory capacity towards CCL21 in mature MUTZ-LCs but not MoLCs was significantly lower when cells were stimulated with S1P. S1P, FTY720 and specific S1P receptor agonists did not modulate the endocytotic capacity of immature MUTZ-LCs and MoLCs. These findings were further supported by testing specific antagonists of S1P1-4 in the absence or presence of S1P. CONCLUSION: Our data demonstrate that S1P regulates key events of human LC maturation including cytokine release and migration. These findings are of particular importance when considering the potential use of S1P in inflammatory skin disorders.


Subject(s)
Cell Differentiation/drug effects , Langerhans Cells/drug effects , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , Cell Line, Tumor , Cell Movement/drug effects , Cytokines/metabolism , Endocytosis/drug effects , Fingolimod Hydrochloride/pharmacology , Humans , Langerhans Cells/immunology , Langerhans Cells/metabolism , Phenotype , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Sphingosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...