Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Science ; 384(6691): eabo7027, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38574142

ABSTRACT

Macrophages are functionally heterogeneous cells essential for apoptotic cell clearance. Apoptotic cells are defined by homogeneous characteristics, ignoring their original cell lineage identity. We found that in an interleukin-4 (IL-4)-enriched environment, the sensing of apoptotic neutrophils by macrophages triggered their tissue remodeling signature. Engulfment of apoptotic hepatocytes promoted a tolerogenic phenotype, whereas phagocytosis of T cells had little effect on IL-4-induced gene expression. In a mouse model of parasite-induced pathology, the transfer of macrophages conditioned with IL-4 and apoptotic neutrophils promoted parasitic egg clearance. Knockout of phagocytic receptors required for the uptake of apoptotic neutrophils and partially T cells, but not hepatocytes, exacerbated helminth infection. These findings suggest that the identity of apoptotic cells may contribute to the development of distinct IL-4-driven immune programs in macrophages.


Subject(s)
Apoptosis , Interleukin-4 , Macrophages , Phagocytosis , Schistosomiasis mansoni , Animals , Mice , Apoptosis/immunology , Hepatocytes/immunology , Interleukin-4/genetics , Interleukin-4/metabolism , Macrophages/immunology , Mice, Knockout , Neutrophils/immunology , Phagocytosis/immunology , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/immunology , Disease Models, Animal
2.
Biomed Pharmacother ; 173: 116286, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401515

ABSTRACT

Lipopolysaccharide (LPS, endotoxin) is ubiquitous and represents a harmful contaminant of pharmaceutical compounds, recombinant biologicals and drug products. The pyrogen can induce severe immune responses and pathology in vitro and in vivo. Health authorities require strict control of endotoxin in parenteral drugs. However, for research and pre-clinical compound analysis, endotoxin testing is not a required quality control, which may cause potential drawbacks in the translational pipeline. Endotoxin testing is usually performed by the Limulus amebocyte lysate (LAL) assay, which is hampered by the so-called low endotoxin recovery (LER) effect when certain drug formulations are tested. A comprehensive study including structural, biophysical, and biological analyses was conducted to identify LER root cause for phosphate- and polysorbate-containing parenteral drug products. LPS in water showed extended ribbon-like aggregate structures. In placebo (formulation buffer without drug) and in drug product (drug in formulation buffer), a reaggregation of LPS into a network of interlinked micelles with hidden head group charges, and a strong reduction of the negative surface potential was observed. The non-accessibility of the LPS backbone has a direct impact leading (i) to a loss of activation of the LAL-cascade, (ii) reduced activation of the TLR4/MD-2 receptor system, and (iii) increased survival in a mouse model of endotoxemia. These data provide a structure-based explanation of the LER-underlying mechanisms. A human whole blood assay is shown to resolve LER and detect the pyrogenic activity of endotoxin with high sensitivity. This may open new test options to improve quality control in drug development and drug safety.


Subject(s)
Endotoxins , Lipopolysaccharides , Animals , Mice , Humans , Micelles , Limulus Test , Drug Compounding
3.
Eur J Pharm Sci ; 175: 106216, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35618202

ABSTRACT

A strong inflammatory immune response drives the lung pathology in neonatal acute respiratory distress syndrome (nARDS). Anti-inflammatory therapy is therefore a promising strategy for improved treatment of nARDS. We demonstrate a new function of the anionic phospholipids POPG, DOPG, and PIP2 as inhibitors of IL-1ß release by LPS and ATP-induced inflammasome activation in human monocyte-derived and lung macrophages. Curosurf® surfactant was enriched with POPG, DOPG, PIP2 and the head-group derivative IP3, biophysically characterized and applicability was evaluated in a piglet model of nARDS. The composition of pulmonary surfactant from piglets was determined by shotgun lipidomics screens. After 72 h of nARDS, levels of POPG, DOPG, and PIP2 were enhanced in the respective treatment groups. Otherwise, we did not observe changes of individual lipid species in any of the groups. Surfactant proteins were not affected, with the exception of the IP3 treated group. Our data show that POPG, DOPG, and PIP2 are potent inhibitors of inflammasome activation; their enrichment in a surfactant preparation did not induce any negative effects on lipid profile and reduced biophysical function in vitro was mainly observed for PIP2. These results encourage to rethink the current strategies of improving surfactant preparations by inclusion of anionic lipids as potent anti-inflammatory immune regulators.


Subject(s)
Pulmonary Surfactants , Respiratory Distress Syndrome , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammasomes/metabolism , Lipidomics , Lung/metabolism , Phospholipids/pharmacology , Pulmonary Surfactants/metabolism , Pulmonary Surfactants/pharmacology , Surface-Active Agents , Swine
4.
EMBO Rep ; 22(7): e52334, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34212478

ABSTRACT

The Research Center Borstel developed a bottom-up approach based on communication and team scouts to create a culture that fosters research integrity.


Subject(s)
Communication , Humans
5.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Article in English | MEDLINE | ID: mdl-34183393

ABSTRACT

Antimicrobial peptides (AMPs) contribute to an effective protection against infections. The antibacterial function of AMPs depends on their interactions with microbial membranes and lipids, such as lipopolysaccharide (LPS; endotoxin). Hyperinflammation induced by endotoxin is a key factor in bacterial sepsis and many other human diseases. Here, we provide a comprehensive profile of peptide-mediated LPS neutralization by systematic analysis of the effects of a set of AMPs and the peptide antibiotic polymyxin B (PMB) on the physicochemistry of endotoxin, macrophage activation, and lethality in mice. Mechanistic studies revealed that the host defense peptide LL-32 and PMB each reduce LPS-mediated activation also via a direct interaction of the peptides with the host cell. As a biophysical basis, we demonstrate modifications of the structure of cholesterol-rich membrane domains and the association of glycosylphosphatidylinositol (GPI)-anchored proteins. Our discovery of a host cell-directed mechanism of immune control contributes an important aspect in the development and therapeutic use of AMPs.


Subject(s)
Cathelicidins/pharmacology , Cell Membrane/metabolism , Host-Pathogen Interactions , Lipopolysaccharides/pharmacology , Neutralization Tests , Polymyxin B/pharmacology , Animals , Antimicrobial Cationic Peptides/pharmacology , Biological Transport/drug effects , Cell Membrane/drug effects , Cholesterol/metabolism , Female , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Inflammation/pathology , Mice, Inbred C57BL , Signal Transduction/drug effects
6.
Expert Rev Anti Infect Ther ; 19(4): 495-517, 2021 04.
Article in English | MEDLINE | ID: mdl-33210958

ABSTRACT

INTRODUCTION: Gram-negative bacterial infections represent still a severe problem of human health care, regarding the increase in multi-resistance against classical antibiotics and the lack of newly developed antimicrobials. For the fight against these germs, anti-infective agents must overcome and/or bind to the Gram-negative outer membrane consisting of a lipopolysaccharide (LPS, endotoxin) outer leaflet and an inner leaflet from phospholipids, with additional peripheral or integral membrane proteins (OMP's). AREAS COVERED: The current article reviews data of existing therapeutic options and summarizes newer approaches for targeting and neutralizing endotoxins, ranging from in vitro over in vivo animal data to clinical applications by using databases such as Medline. EXPERT OPINION: Conventional antibiotic treatment of the bacteria leads to their killing, but not necessary LPS neutralization, which may be a severe problem in particular for the systemic pathway. This is the reason why there is an increasing number of therapeutic approaches, which - besides combating whole bacteria - at the same time try to neutralize endotoxin within or outside the bacterial cells mainly responsible for the high inflammation induction in Gram-negative species.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Drug Development , Endotoxins/antagonists & inhibitors , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacterial Infections/microbiology , Humans , Inflammation/drug therapy , Inflammation/microbiology , Lipopolysaccharides/antagonists & inhibitors
7.
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
8.
Front Immunol ; 10: 122, 2019.
Article in English | MEDLINE | ID: mdl-30837983

ABSTRACT

Molecular allergology research has provided valuable information on the structure and function of single allergenic molecules. There are several allergens in food and inhalant allergen sources that are able to interact with lipid ligands via different structural features: hydrophobic pockets, hydrophobic cavities, or specialized domains. For only a few of these allergens information on their associated ligands is already available. Several of the allergens are clinically relevant, so that it is highly probable that the individual structural features with which they interact with lipids have a direct effect on their allergenic potential, and thus on allergy development. There is some evidence for a protective effect of lipids delaying the enzymatic digestion of the peanut (Arachis hypogaea) allergen Ara h 8 (hydrophobic pocket), probably allowing this molecule to get to the intestinal immune system intact (sensitization). Oleosins from different food allergen sources are part of lipid storage organelles and potential marker allergens for the severity of the allergic reaction. House dust mite (HDM), is more often associated with allergic asthma than other sources of inhalant allergens. In particular, lipid-associated allergens from Dermatophagoides pteronyssinus which are Der p 2, Der p 5, Der p 7, Der p 13, Der p 14, and Der p 21 have been reported to be associated with severe allergic reactions and respiratory symptoms such as asthma. The exact mechanism of interaction of these allergens with lipids still has to be elucidated. Apart from single allergens glycolipids have been shown to directly induce allergic inflammation. Several-in parts conflicting-data exist on the lipid (and allergen) and toll-like receptor interactions. For only few single allergens mechanistic studies were performed on their interaction with the air-liquid interface of the lungs, in particular with the surfactant components SP-A and SP-D. The increasing knowledge on protein-lipid-interaction for lipophilic and hydrophobic food and inhalant allergens on the basis of their particular structure, of their capacity to be integral part of membranes (like the oleosins), and their ability to interact with membranes, surfactant components, and transport lipids (like the lipid transfer proteins) are essential to eventually clarify allergy and asthma development.


Subject(s)
Allergens/metabolism , Antigens, Plant/metabolism , Asthma/immunology , Carrier Proteins/metabolism , Hypersensitivity/immunology , Lipids/immunology , Plant Proteins/metabolism , Allergens/immunology , Animals , Antigens, Plant/immunology , Carrier Proteins/immunology , Humans , Lipid Metabolism , Plant Proteins/immunology , Plants , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein D/immunology , Pulmonary Surfactant-Associated Protein D/metabolism
9.
FEBS J ; 286(8): 1576-1593, 2019 04.
Article in English | MEDLINE | ID: mdl-30843356

ABSTRACT

Increasing failure of conventional antibiotics to combat bacterial infections requires the urgent development of new antibacterial drugs; a promising class of new drugs based on antimicrobial peptides. Here, we studied the molecular interaction of polycationic synthetic antilipopolysaccharide peptides (SALPs) with various gram-negative and gram-positive bacteria, including resistant strains. The analysis of antimicrobial activity by conventional techniques and atomic force microscopy showed a strict dependence on amino acid (aa) sequences, with the type of amino acid, its position within the primary structure, and the sequence length being critical parameters. By monitoring lipopolysaccharide (LPS)- or bacteria-induced cytokine production in human mononuclear cells and whole blood, we found a direct link between the binding of the lead compound Pep19-2.5 to Salmonella enterica and the anti-inflammatory activity of the peptide. Thermodynamic analysis of Pep19-2.5 binding to the bacterial cell envelope showed an exothermic reaction with saturation characteristics, whereas small-angle X-ray scattering data indicated a direct attachment of Pep19-2.5 to the bacterial cell envelope. This binding preferentially takes place to the LPS outer monolayer, as evidenced by the change in the LPS acyl chain and phosphate vibrational bands seen by Fourier-transform infrared spectroscopy. We report here that the anti-inflammatory activity of Pep19-2.5 is not only connected with neutralization of cell-free bacterial toxins but also with a direct binding of the peptide to the outer leaflet of the bacterial outer membrane.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/metabolism , Peptides/metabolism , Peptides/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Calorimetry , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/microbiology , Cesium Radioisotopes/toxicity , Cytokines/metabolism , Flow Cytometry , Humans , Hydrophobic and Hydrophilic Interactions , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Lipopolysaccharides/pharmacology , Microbial Sensitivity Tests , Microscopy, Atomic Force , Peptides/chemical synthesis , Salmonella enterica/drug effects , Salmonella enterica/metabolism , Salmonella enterica/radiation effects , Scattering, Small Angle , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
10.
Int J Chron Obstruct Pulmon Dis ; 13: 2255-2259, 2018.
Article in English | MEDLINE | ID: mdl-30100715

ABSTRACT

Introduction: As part of a study aimed at illuminating at least some of the complex molecular events taking place in COPD, we screened tissues by means of transcriptome analyses. Materials and methods: Tissues were subjected to transcriptome analysis. Candidate genes were identified and validated by immunohistochemistry. Primary human lung cells were subjected to stimulation with cigarette smoke extract for further validation by real time PCR. Results: Six candidate genes were selected for further investigations: Aquaporin 3 (AQP3), extracellular matrix protein 1 (ECM1), four and a half LIM domain 1 (FHL1), milk fat globule epidermal growth factor 8 (MFGE8, lactadherin), phosphodiesterase 4D-interacting protein (PDE4DIP), and creatine transporter SLC6A8. All six proteins were allocated to distinct cell types by immunohistochemistry. Upon stimulation with cigarette smoke extract, human type II pneumocytes showed a dose-dependent down-regulation of MFGE8, while ECM1 and FHL1 also tended to be down-regulated. Although present, none of the candidates was regulated by cigarette smoke extract in primary human macrophages. Discussion: MFGE8 turned out to be an interesting new candidate gene in COPD deserving further studies.


Subject(s)
Antigens, Surface/genetics , Aquaporin 3/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Profiling/methods , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Milk Proteins/genetics , Muscle Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Adaptor Proteins, Signal Transducing , Aged , Cytoskeletal Proteins , Down-Regulation , Female , Germany , Humans , Lung , Male , Real-Time Polymerase Chain Reaction , Smoke
11.
J Med Chem ; 61(7): 2895-2909, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29494148

ABSTRACT

The structure-activity relationship was investigated in a series of synthetic TLR4 antagonists formed by a glucosamine core linked to two phosphate esters and two linear carbon chains. Molecular modeling showed that the compounds with 10, 12, and 14 carbons chains are associated with higher stabilization of the MD-2/TLR4 antagonist conformation than in the case of the C16 variant. Binding experiments with human MD-2 showed that the C12 and C14 variants have higher affinity than C10, while the C16 variant did not interact with the protein. The molecules, with the exception of the C16 variant, inhibited the LPS-stimulated TLR4 signal in human and murine cells, and the antagonist potency mirrored the MD-2 affinity calculated from in vitro binding experiments. Fourier-transform infrared, nuclear magnetic resonance, and small angle X-ray scattering measurements suggested that the aggregation state in aqueous solution depends on fatty acid chain lengths and that this property can influence TLR4 activity in this series of compounds.


Subject(s)
Monosaccharides/chemistry , Monosaccharides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Binding, Competitive/drug effects , Cell Line , Fatty Acids/chemistry , HEK293 Cells , Humans , Interleukin-8/biosynthesis , Ligands , Lipopolysaccharides/metabolism , Mice , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship
12.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L32-L53, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28860142

ABSTRACT

The biological and immune-protective properties of surfactant-derived phospholipids and phospholipid subfractions in the context of neonatal inflammatory lung disease are widely unknown. Using a porcine neonatal triple-hit acute respiratory distress syndrome (ARDS) model (repeated airway lavage, overventilation, and LPS instillation into airways), we assessed whether the supplementation of surfactant (S; poractant alfa) with inositol derivatives [inositol 1,2,6-trisphosphate (IP3) or phosphatidylinositol 3,5-bisphosphate (PIP2)] or phosphatidylglycerol subfractions [16:0/18:1-palmitoyloleoyl-phosphatidylglycerol (POPG) or 18:1/18:1-dioleoyl-phosphatidylglycerol (DOPG)] would result in improved clinical parameters and sought to characterize changes in key inflammatory pathways behind these improvements. Within 72 h of mechanical ventilation, the oxygenation index (S+IP3, S+PIP2, and S+POPG), the ventilation efficiency index (S+IP3 and S+POPG), the compliance (S+IP3 and S+POPG) and resistance (S+POPG) of the respiratory system, and the extravascular lung water index (S+IP3 and S+POPG) significantly improved compared with S treatment alone. The inositol derivatives (mainly S+IP3) exerted their actions by suppressing acid sphingomyelinase activity and dependent ceramide production, linked with the suppression of the inflammasome nucleotide-binding domain, leucine-rich repeat-containing protein-3 (NLRP3)-apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-caspase-1 complex, and the profibrotic response represented by the cytokines transforming growth factor-ß1 and IFN-γ, matrix metalloproteinase (MMP)-1/8, and elastin. In addition, IκB kinase activity was significantly reduced. S+POPG and S+DOPG treatment inhibited polymorphonuclear leukocyte activity (MMP-8 and myeloperoxidase) and the production of interleukin-6, maintained alveolar-capillary barrier functions, and reduced alveolar epithelial cell apoptosis, all of which resulted in reduced pulmonary edema. S+DOPG also limited the profibrotic response. We conclude that highly concentrated inositol derivatives and phosphatidylglycerol subfractions in surfactant preparations mitigate key inflammatory pathways in inflammatory lung disease and that their clinical application may be of interest for future treatment of the acute exudative phase of neonatal ARDS.


Subject(s)
Disease Models, Animal , Inositol/pharmacology , Phosphatidylglycerols/pharmacology , Pulmonary Edema/drug therapy , Pulmonary Surfactants/pharmacology , Respiratory Distress Syndrome, Newborn/drug therapy , Animals , Animals, Newborn , Apoptosis , Bronchoalveolar Lavage Fluid , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Male , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Gas Exchange , Random Allocation , Respiration, Artificial , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Swine , Translational Research, Biomedical , Vitamin B Complex/pharmacology
13.
Biochim Biophys Acta ; 1863(4): 660-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26804480

ABSTRACT

The lipopolysaccharide-binding protein (LBP) is critically involved in innate immune responses to Gram-negative infections. We show here that human peripheral blood-derived monocytes, but not lymphocytes, stain positive for endogenous LBP on the cell surface. Studies on human macrophages demonstrate LBP binding at normal serum concentrations of 1-10 µg/ml. Binding was increased in a concentration-dependent manner by lipopolysaccharide (LPS). Fluorescence quenching experiments and confocal microscopy revealed constitutive and LPS-induced internalization of LBP by macrophages. Experiments with macrophages and HEK293 cell lines showed that binding and uptake of LBP do not depend on the LPS receptors CD14 and TLR4/MD-2. Fractionation of Triton X-100 solubilized cytoplasmic membranes revealed that LBP was primarily localized in non-raft domains under resting conditions. Cellular LPS stimulation elevated LBP levels and induced enrichment in fractions marking the transition between non-raft and raft domains. LBP was found to colocalize with LPS at the cytoplasmic membrane and in intracellular compartments of macrophages. In macrophages stimulated with LPS and ATP for inflammasome activation, LBP was observed in close vicinity to activated caspases. Furthermore, LBP conferred IL-1ß production by LPS in the absence of ATP. These data establish that LBP serves not only as an extracellular LPS shuttle but in addition facilitates intracellular transport of LPS. This observation adds a new function to this central immune regulator of LPS biology and raises the possibility for a role of LBP in the delivery of LPS to TLR4-independent intracellular receptors.


Subject(s)
Acute-Phase Proteins/metabolism , Carrier Proteins/metabolism , Cytoplasm/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins/metabolism , Biological Transport , Cells, Cultured , Endocytosis , HEK293 Cells , Humans , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Protein Binding , Signal Transduction/physiology , Tissue Distribution , Toll-Like Receptor 4/metabolism
14.
Cell Microbiol ; 17(5): 607-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25363599

ABSTRACT

The formation and release of outer membrane vesicles (OMVs) is a phenomenon observed in many bacteria, including Legionella pneumophila. During infection, this human pathogen primarily invades alveolar macrophages and replicates within a unique membrane-bound compartment termed Legionella-containing vacuole. In the current study, we analysed the membrane architecture of L. pneumophila OMVs by small-angle X-ray scattering and biophysically characterized OMV membranes. We investigated the interaction of L. pneumophila OMVs with model membranes by Förster resonance energy transfer and Fourier transform infrared spectroscopy. These experiments demonstrated the incorporation of OMV membrane material into liposomes composed of different eukaryotic phospholipids, revealing an endogenous property of OMVs to fuse with eukaryotic membranes. Cellular co-incubation experiments showed a dose- and time-dependent binding of fluorophore-labelled OMVs to macrophages. Trypan blue quenching experiments disclosed a rapid internalization of OMVs into macrophages at 37 and 4 °C. Purified OMVs induced tumour necrosis factor-α production in human macrophages at concentrations starting at 300 ng ml(-1). Experiments on HEK293-TLR2 and TLR4/MD-2 cell lines demonstrated a dominance of TLR2-dependent signalling pathways. In summary, we demonstrate binding, internalization and biological activity of L. pneumophila OMVs on human macrophages. Our data support OMV membrane fusion as a mechanism for the remote delivery of virulence factors to host cells.


Subject(s)
Cell Membrane/metabolism , Exosomes/metabolism , Host-Pathogen Interactions , Legionella pneumophila/physiology , Virulence Factors/metabolism , Biophysical Phenomena , Cells, Cultured , Endocytosis , Epithelial Cells/metabolism , Exosomes/chemistry , Fluorescence Resonance Energy Transfer , Humans , Legionella pneumophila/cytology , Macrophages/immunology , Macrophages/metabolism , Scattering, Small Angle , Spectroscopy, Fourier Transform Infrared , Temperature , Tumor Necrosis Factor-alpha/metabolism
15.
Innate Immun ; 20(8): 787-98, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24122298

ABSTRACT

The lung is constantly exposed to immune stimulation by LPS from inhaled microorganisms. A primary mechanism to maintain immune homeostasis is based on anti-inflammatory regulation by surfactant protein A (SP-A), a secreted component of lung innate immunity. The architecture of LPS aggregates is strongly associated with biological activity. We therefore investigated whether SP-A affects the physico-chemical properties of LPS. Determination of the three-dimensional aggregate structure of LPS by small-angle X-ray scattering demonstrated that SP-A induced the formation of multi-lamellar aggregate structures. Determination of the acyl-chain-fluidity of LPS aggregates by Fourier transform infrared (FTIR) spectroscopy showed that the phase transition temperature of LPS was reduced in the presence of SP-A. The phosphate groups at the diglucosamine backbone of LPS represent important functional groups for the bioactivity of LPS. FTIR analysis revealed changes in the vibrational bands νas PO-(2), indicating an interaction of SP-A with the 1-phosphate, but not with the 4'-phosphate. The physico-chemical changes induced by SP-A were associated with up to 90% reduction in LPS-induced TNF-α-production by human macrophages. In conclusion, our data demonstrate that the SP-A/LPS interaction induces conformational changes in LPS aggregates leading to biologically less active structures, thereby providing a new molecular mechanism of immune modulation by SP-A.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/drug effects , Pulmonary Surfactant-Associated Protein A/pharmacology , HEK293 Cells , Humans , In Vitro Techniques , Lipopolysaccharides/chemistry , Macrophage Activation/drug effects , Molecular Conformation/drug effects , Protein Conformation , Tumor Necrosis Factor-alpha/metabolism
16.
FEBS J ; 280(4): 1095-111, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23279861

ABSTRACT

We established a new preparative separation procedure, based on DOC/PAGE, to isolate intact lipopolysaccharide (LPS) fractions from natural LPS preparations of Escherichia coli. Analysis of the chemical integrity of LPS fractions by MS showed that no significant chemical modifications were introduced by the procedure. Contamination with toll-like receptor 2 (TLR2)-reactive cell-wall components present in the natural LPS mixture was effectively removed by the procedure, as determined by the absence of reactivity of the purified fractions in a HEK293-TLR2 cell line. Biologic analysis of LPS fractions derived from E. coli O111 in human macrophages demonstrated that the rough (R), semirough (SR) and smooth (S) LPS fractions were highly active at inducing tumor necrosis factor-alpha (TNF-α) in the presence of human serum; however, on a weight basis the R-LPS and SR-LPS fractions were more active, by a factor of 10-100, than was the S-LPS fraction. Under serum-free conditions, the natural LPS mixture, as well as the R-LPS and SR-LPS fractions, showed dose-dependent activation of macrophages, although the response was attenuated by about 10- to 100-fold. In contrast, the S-LPS fraction failed to induce TNF-α. Remarkably, the dose-response of the natural LPS mixture resembled that of the R-LPS and SR-LPS fractions, supporting that short-chain (R and SR) forms of LPS dominate the innate immune response of human macrophages to LPS in vitro. Biologic activity to the S-LPS fraction under serum-free conditions could be restored by the addition of recombinant lipopolysaccharide-binding protein (LBP). In contrast, soluble cluster of differentiation antigen 14 was not able to confer activity of the S-LPS fraction, indicating a crucial role of LBP in the recognition of S-LPS by human macrophages.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/immunology , Shiga-Toxigenic Escherichia coli/chemistry , Chromatography, Reverse-Phase , Deoxycholic Acid/chemistry , Electrophoresis, Polyacrylamide Gel , Fourier Analysis , Glycosylation , HEK293 Cells , Humans , Lipopolysaccharides/chemistry , Lipopolysaccharides/isolation & purification , Macrophage Activation , Macrophages/metabolism , Spectrometry, Mass, Electrospray Ionization , Toll-Like Receptor 2/agonists , Tumor Necrosis Factor-alpha/metabolism
17.
Adv Exp Med Biol ; 667: 39-51, 2010.
Article in English | MEDLINE | ID: mdl-20665198

ABSTRACT

Entry of endotoxin (lipopolysaccharide (LPS) or lipid A) into the blood stream is causative for the emergence of sepsis and septic shock with all its pathophysiological consequences.(1) Serum contains a whole variety of proteins that interact with endotoxin. As large as the number of different proteins interacting with endotoxin, as broad are the consequences of these interactions. Serum proteins can either enhance cell activation by endotoxin or attenuate the cellular response, they can detoxify and eliminate endotoxin from the blood stream. In this chapter we summarize work on the investigation of the interaction of endotoxins with serum proteins. In four paragraphs we focus on proteins involved in the endotoxin-induced immune cell activation, detection by immunoglobulins, the transport of endotoxins and on proteins and peptides with the capability to neutralize the biological effects of endotoxin. There is a multitude of studies analyzing the interactions between serum proteins and endotoxins, however, with great differences in the source and quality of the endotoxins used. The number of studies dealing with chemically well defined endotoxin structures are quite limited. In addition, though lipid A is the biologically active entity, the "endotoxic principle", of LPS, the majority of studies was performed with LPS. Therefore, to be comprehensive, we included also studies dealing with LPS and not with lipid A if fundamental scientific problems were addressed. In that cases, we have to be aware that there may be differences in the protein interactions of lipid A and LPS, and we tried to emphasize this point in the respective paragraphs.


Subject(s)
Blood Proteins/metabolism , Lipid A/metabolism , Amino Acid Sequence , Blood Proteins/chemistry , Humans , Immunoglobulins/metabolism , Molecular Sequence Data
18.
Innate Immun ; 16(4): 213-25, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19710101

ABSTRACT

The innate immune response provides a critical first-line defense against Mycobacterium tuberculosis, an intracellular pathogen that represents a major health threat world-wide. A synthetic lipopeptide (LP) mimicking the lipid moiety of the cell-wall associated 19-kDa lipoprotein from M. tuberculosis has recently been assigned an important role in the induction of an antibacterial immune response in host macrophages. Here, we present experimental data on the biological activities and the biophysical mechanisms underlying cell activation by synthetic 19-kDa M. tuberculosis-derived lipopeptide (Mtb-LP). Investigation of the geometry of the LP (i.e. the molecular conformation and supramolecular aggregate structure) and the preference for membrane intercalation provide an explanation for the biological activities of the mycobacterial LP. Cell activation by low concentrations of Mtb-LP was enhanced by the lipopolysaccharide-binding protein and CD14. However, surprisingly, we found that activation of human macrophages to induce pro- as well as antiinflammatory mediators (tumor necrosis factor(TNF)-alpha, Interleukin(IL)-6, IL-8, and IL-10) in response to the Mtb-LP is strongly reduced in the presence of serum. This observation could be confirmed for the immune response of murine macrophages which showed a strongly enhanced TNF-alpha release in the absence of serum, suggesting that the molecular mechanisms of immune recognition of the Mtb-LP are tailored to the ambient conditions of the lung.


Subject(s)
Bacterial Proteins/immunology , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/immunology , Serum/immunology , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Biomimetics , Bone Marrow Cells/immunology , Culture Media, Serum-Free , Cytokines/analysis , Cytokines/biosynthesis , Fluorescence Resonance Energy Transfer , Humans , Immunity, Cellular/immunology , Immunity, Innate/immunology , Indicators and Reagents , Inflammation/immunology , Lipids/chemistry , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
19.
J Immunol ; 179(7): 4480-91, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17878344

ABSTRACT

The pulmonary collectin surfactant protein (SP)-A has a pivotal role in anti-inflammatory modulation of lung immunity. The mechanisms underlying SP-A-mediated inhibition of LPS-induced NF-kappaB activation in vivo and in vitro are only partially understood. We previously demonstrated that SP-A stabilizes IkappaB-alpha, the primary regulator of NF-kappaB, in alveolar macrophages (AM) both constitutively and in the presence of LPS. In this study, we show that in AM and PBMC from IkappaB-alpha knockout/IkappaB-beta knockin mice, SP-A fails to inhibit LPS-induced TNF-alpha production and p65 nuclear translocation, confirming a critical role for IkappaB-alpha in SP-A-mediated LPS inhibition. We identify atypical (a) protein kinase C (PKC) zeta as a pivotal upstream regulator of SP-A-mediated IkappaB-alpha/NF-kappaB pathway modulation deduced from blocking experiments and confirmed by using AM from PKCzeta-/- mice. SP-A transiently triggers aPKCThr(410/403) phosphorylation, aPKC kinase activity, and translocation in primary rat AM. Coimmunoprecipitation experiments reveal that SP-A induces aPKC/p65 binding under constitutive conditions. Together the data indicate that anti-inflammatory macrophage activation via IkappaB-alpha by SP-A critically depends on PKCzeta activity, and thus attribute a novel, stimulus-specific signaling function to PKCzeta in SP-A-modulated pulmonary immune response.


Subject(s)
I-kappa B Kinase/metabolism , Protein Kinase C/metabolism , Pulmonary Surfactant-Associated Protein A/therapeutic use , Active Transport, Cell Nucleus , Animals , Cell Membrane/enzymology , Cells, Cultured , Enzyme Activation , I-kappa B Kinase/genetics , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Transgenic , Mutation/genetics , NF-kappa B/metabolism , Phosphothreonine/metabolism , Protein Binding , Protein Kinase C/deficiency , Protein Kinase C/genetics , Rats , Tumor Necrosis Factor-alpha/metabolism
20.
Int J Med Microbiol ; 297(5): 341-52, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17452014

ABSTRACT

It is generally accepted that the interaction of bacterial pathogenicity factors such as endotoxin (lipopolysaccharide, LPS) with molecules and cells of the human immune system, which eventually may lead to pathophysiological effects like septic shock, is exerted by isolated molecules after their release from the bacteria. Therefore, the study of the direct, physical interaction of LPS with target structures by applying biophysical means is of high interest. The questions which arise in this context concern the biologically active unit of LPS (monomer, multimer, aggregate), the molecular conformation of the single molecules, the type of aggregation of LPS polymers, the strength of their binding to serum and membrane proteins and/or unspecific binding to membrane phospholipids. Here, recent progress is reviewed which has increased our understanding of the processes preceding LPS-induced immune cell activation.


Subject(s)
Endotoxins/chemistry , Endotoxins/toxicity , Lipopolysaccharides/metabolism , Lipopolysaccharides/toxicity , Receptors, Immunologic/metabolism , Bacterial Infections/complications , Humans , Lipopolysaccharides/immunology , Shock, Septic/immunology , Shock, Septic/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...