Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
PLoS One ; 17(12): e0276115, 2022.
Article in English | MEDLINE | ID: mdl-36538516

ABSTRACT

Human-based organ models can provide strong predictive value to investigate the tropism, virulence, and replication kinetics of viral pathogens. Currently, such models have received widespread attention in the study of SARS-CoV-2 causing the COVID-19 pandemic. Applicable to a large set of organoid models and viruses, we provide a step-by-step work instruction for the infection of human alveolar-like organoids with SARS-CoV-2 in this protocol collection. We also prepared a detailed description on state-of-the-art methodologies to assess the infection impact and the analysis of relevant host factors in organoids. This protocol collection consists of five different sets of protocols. Set 1 describes the protein extraction from human alveolar-like organoids and the determination of protein expression of angiotensin-converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2) and FURIN as exemplary host factors of SARS-CoV-2. Set 2 provides detailed guidance on the extraction of RNA from human alveolar-like organoids and the subsequent qPCR to quantify the expression level of ACE2, TMPRSS2, and FURIN as host factors of SARS-CoV-2 on the mRNA level. Protocol set 3 contains an in-depth explanation on how to infect human alveolar-like organoids with SARS-CoV-2 and how to quantify the viral replication by plaque assay and viral E gene-based RT-qPCR. Set 4 provides a step-by-step protocol for the isolation of single cells from infected human alveolar-like organoids for further processing in single-cell RNA sequencing or flow cytometry. Set 5 presents a detailed protocol on how to perform the fixation of human alveolar-like organoids and guides through all steps of immunohistochemistry and in situ hybridization to visualize SARS-CoV-2 and its host factors. The infection and all subsequent analytical methods have been successfully validated by biological replications with human alveolar-like organoids based on material from different donors.


Subject(s)
COVID-19 , Humans , COVID-19/metabolism , SARS-CoV-2 , Furin/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Pandemics , Lung/metabolism , Organoids
3.
Eur Respir J ; 60(6)2022 12.
Article in English | MEDLINE | ID: mdl-35728978

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilises the angiotensin-converting enzyme 2 (ACE2) transmembrane peptidase as cellular entry receptor. However, whether SARS-CoV-2 in the alveolar compartment is strictly ACE2-dependent and to what extent virus-induced tissue damage and/or direct immune activation determines early pathogenesis is still elusive. METHODS: Spectral microscopy, single-cell/-nucleus RNA sequencing or ACE2 "gain-of-function" experiments were applied to infected human lung explants and adult stem cell derived human lung organoids to correlate ACE2 and related host factors with SARS-CoV-2 tropism, propagation, virulence and immune activation compared to SARS-CoV, influenza and Middle East respiratory syndrome coronavirus (MERS-CoV). Coronavirus disease 2019 (COVID-19) autopsy material was used to validate ex vivo results. RESULTS: We provide evidence that alveolar ACE2 expression must be considered scarce, thereby limiting SARS-CoV-2 propagation and virus-induced tissue damage in the human alveolus. Instead, ex vivo infected human lungs and COVID-19 autopsy samples showed that alveolar macrophages were frequently positive for SARS-CoV-2. Single-cell/-nucleus transcriptomics further revealed nonproductive virus uptake and a related inflammatory and anti-viral activation, especially in "inflammatory alveolar macrophages", comparable to those induced by SARS-CoV and MERS-CoV, but different from NL63 or influenza virus infection. CONCLUSIONS: Collectively, our findings indicate that severe lung injury in COVID-19 probably results from a macrophage-triggered immune activation rather than direct viral damage of the alveolar compartment.


Subject(s)
COVID-19 , Influenza, Human , Adult , Humans , Angiotensin-Converting Enzyme 2 , Lung/pathology , Macrophages, Alveolar/metabolism , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2 , Viral Tropism
4.
Anesthesiology ; 132(4): 795-807, 2020 04.
Article in English | MEDLINE | ID: mdl-32101978

ABSTRACT

BACKGROUND: Community-acquired pneumonia and associated sepsis cause high mortality despite antibiotic treatment. Uncontrolled inflammatory host responses contribute to the unfavorable outcome by driving lung and extrapulmonary organ failure. The complement fragment C5a holds significant proinflammatory functions and is associated with tissue damage in various inflammatory conditions. The authors hypothesized that C5a concentrations are increased in pneumonia and C5a neutralization promotes barrier stabilization in the lung and is protective in pneumococcal pulmonary sepsis. METHODS: The authors investigated regulation of C5a in pneumonia in a prospective patient cohort and in experimental pneumonia. Two complementary models of murine pneumococcal pneumonia were applied. Female mice were treated with NOX-D19, a C5a-neutralizing L-RNA-aptamer. Lung, liver, and kidney injury and the inflammatory response were assessed by measuring pulmonary permeability (primary outcome), pulmonary and blood leukocytes, cytokine concentrations in lung and blood, and bacterial load in lung, spleen, and blood, and performing histologic analyses of tissue damage, apoptosis, and fibrin deposition (n = 5 to 13). RESULTS: In hospitalized patients with pneumonia (n = 395), higher serum C5a concentrations were observed compared to healthy subjects (n = 24; 6.3 nmol/l [3.9 to 10.0] vs. 4.5 nmol/l [3.8 to 6.6], median [25 to 75% interquartile range]; difference: 1.4 [95% CI, 0.1 to 2.9]; P = 0.029). Neutralization of C5a in mice resulted in lower pulmonary permeability in pneumococcal pneumonia (1.38 ± 0.89 vs. 3.29 ± 2.34, mean ± SD; difference: 1.90 [95% CI, 0.15 to 3.66]; P = 0.035; n = 10 or 11) or combined severe pneumonia and mechanical ventilation (2.56 ± 1.17 vs. 7.31 ± 5.22; difference: 4.76 [95% CI, 1.22 to 8.30]; P = 0.011; n = 9 or 10). Further, C5a neutralization led to lower blood granulocyte colony-stimulating factor concentrations and protected against sepsis-associated liver injury. CONCLUSIONS: Systemic C5a is elevated in pneumonia patients. Neutralizing C5a protected against lung and liver injury in pneumococcal pneumonia in mice. Early neutralization of C5a might be a promising adjunctive treatment strategy to improve outcome in community-acquired pneumonia.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Complement C5a/antagonists & inhibitors , Pneumonia, Pneumococcal/blood , Pneumonia, Pneumococcal/prevention & control , Sepsis/blood , Sepsis/prevention & control , Animals , Antibodies, Neutralizing/administration & dosage , Biomarkers/blood , Cohort Studies , Complement C5a/metabolism , Female , Immunologic Factors/antagonists & inhibitors , Immunologic Factors/blood , Mice , Mice, Inbred C57BL , Prospective Studies
6.
Z Kinder Jugendpsychiatr Psychother ; 46(2): 123-132, 2018 Mar.
Article in German | MEDLINE | ID: mdl-28165300

ABSTRACT

Objective: A descriptive analysis of victim compensation applications for children and adolescents as well as sociodemographic and trauma-specific information concerning victims and perpetrators. Method: We did analysis of 100 victim-compensation application files based on a self-developed category system. Results: The files included solely interpersonal trauma, 59 % of which are type II trauma. The most frequent form is sexual violence. The perpetrators stem mostly from children's homes or peripherals. 79 % of the victims received a diagnosis of a mental disorder, most often posttraumatic stress disorder. Conclusions: Sexually abused children and adolescents make up the majority of the target population in OEG-related trauma outpatient units. Such outpatient units should therefore offer a specific expertise in treating sexually abused children and adolescents.


Subject(s)
Child Abuse/legislation & jurisprudence , Compensation and Redress/legislation & jurisprudence , Crime Victims/legislation & jurisprudence , Exposure to Violence/legislation & jurisprudence , Life Change Events , Minors/legislation & jurisprudence , Survivors/legislation & jurisprudence , Adolescent , Child , Child Abuse, Sexual/legislation & jurisprudence , Child, Preschool , Crime Victims/psychology , Female , Germany , Humans , Infant , Male , Minors/psychology , Stress Disorders, Post-Traumatic/diagnosis , Stress Disorders, Post-Traumatic/psychology
7.
Crit Care ; 19: 23, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25888164

ABSTRACT

INTRODUCTION: Lung-protective ventilation reduced acute respiratory distress syndrome (ARDS) mortality. To minimize ventilator-induced lung injury (VILI), tidal volume is limited, high plateau pressures are avoided, and positive end-expiratory pressure (PEEP) is applied. However, the impact of specific ventilatory patterns on VILI is not well defined. Increasing inspiratory time and thereby the inspiratory/expiratory ratio (I:E ratio) may improve oxygenation, but may also be harmful as the absolute stress and strain over time increase. We thus hypothesized that increasing inspiratory time and I:E ratio aggravates VILI. METHODS: VILI was induced in mice by high tidal-volume ventilation (HVT 34 ml/kg). Low tidal-volume ventilation (LVT 9 ml/kg) was used in control groups. PEEP was set to 2 cm H2O, FiO2 was 0.5 in all groups. HVT and LVT mice were ventilated with either I:E of 1:2 (LVT 1:2, HVT 1:2) or 1:1 (LVT 1:1, HVT 1:1) for 4 hours or until an alternative end point, defined as mean arterial blood pressure below 40 mm Hg. Dynamic hyperinflation due to the increased I:E ratio was excluded in a separate group of animals. Survival, lung compliance, oxygenation, pulmonary permeability, markers of pulmonary and systemic inflammation (leukocyte differentiation in lung and blood, analyses of pulmonary interleukin-6, interleukin-1ß, keratinocyte-derived chemokine, monocyte chemoattractant protein-1), and histopathologic pulmonary changes were analyzed. RESULTS: LVT 1:2 or LVT 1:1 did not result in VILI, and all individuals survived the ventilation period. HVT 1:2 decreased lung compliance, increased pulmonary neutrophils and cytokine expression, and evoked marked histologic signs of lung injury. All animals survived. HVT 1:1 caused further significant worsening of oxygenation, compliance and increased pulmonary proinflammatory cytokine expression, and pulmonary and blood neutrophils. In the HVT 1:1 group, significant mortality during mechanical ventilation was observed. CONCLUSION: According to the "baby lung" concept, mechanical ventilation-associated stress and strain in overinflated regions of ARDS lungs was simulated by using high tidal-volume ventilation. Increase of inspiratory time and I:E ratio significantly aggravated VILI in mice, suggesting an impact of a "stress/strain × time product" for the pathogenesis of VILI. Thus increasing the inspiratory time and I:E ratio should be critically considered.


Subject(s)
Exhalation , Inhalation , Lung/pathology , Respiration, Artificial/adverse effects , Tidal Volume , Ventilator-Induced Lung Injury/physiopathology , Animals , Female , Mice , Mice, Inbred C57BL , Respiration, Artificial/methods , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/physiopathology , Ventilator-Induced Lung Injury/complications , Ventilator-Induced Lung Injury/pathology
8.
Crit Care ; 18(2): R73, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24731244

ABSTRACT

INTRODUCTION: Ventilator-induced lung injury (VILI) contributes to morbidity and mortality in acute respiratory distress syndrome (ARDS). Particularly pre-injured lungs are susceptible to VILI despite protective ventilation. In a previous study, the endogenous peptide adrenomedullin (AM) protected murine lungs from VILI. We hypothesized that mechanical ventilation (MV) contributes to lung injury and sepsis in pneumonia, and that AM may reduce lung injury and multiple organ failure in ventilated mice with pneumococcal pneumonia. METHODS: We analyzed in mice the impact of MV in established pneumonia on lung injury, inflammation, bacterial burden, hemodynamics and extrapulmonary organ injury, and assessed the therapeutic potential of AM by starting treatment at intubation. RESULTS: In pneumococcal pneumonia, MV increased lung permeability, and worsened lung mechanics and oxygenation failure. MV dramatically increased lung and blood cytokines but not lung leukocyte counts in pneumonia. MV induced systemic leukocytopenia and liver, gut and kidney injury in mice with pneumonia. Lung and blood bacterial burden was not affected by MV pneumonia and MV increased lung AM expression, whereas receptor activity modifying protein (RAMP) 1-3 expression was increased in pneumonia and reduced by MV. Infusion of AM protected against MV-induced lung injury (66% reduction of pulmonary permeability p < 0.01; prevention of pulmonary restriction) and against VILI-induced liver and gut injury in pneumonia (91% reduction of AST levels p < 0.05, 96% reduction of alanine aminotransaminase (ALT) levels p < 0.05, abrogation of histopathological changes and parenchymal apoptosis in liver and gut). CONCLUSIONS: MV paved the way for the progression of pneumonia towards ARDS and sepsis by aggravating lung injury and systemic hyperinflammation leading to liver, kidney and gut injury. AM may be a promising therapeutic option to protect against development of lung injury, sepsis and extrapulmonary organ injury in mechanically ventilated individuals with severe pneumonia.


Subject(s)
Adrenomedullin/therapeutic use , Bronchodilator Agents/therapeutic use , Pneumonia, Pneumococcal/prevention & control , Respiration, Artificial/adverse effects , Sepsis/prevention & control , Ventilator-Induced Lung Injury/prevention & control , Animals , Female , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/pathology , Sepsis/pathology , Ventilator-Induced Lung Injury/pathology
9.
PLoS One ; 7(5): e35832, 2012.
Article in English | MEDLINE | ID: mdl-22563471

ABSTRACT

BACKGROUND: Even protective ventilation may aggravate or induce lung failure, particularly in preinjured lungs. Thus, new adjuvant pharmacologic strategies are needed to minimize ventilator-induced lung injury (VILI). Intermedin/Adrenomedullin-2 (IMD) stabilized pulmonary endothelial barrier function in vitro. We hypothesized that IMD may attenuate VILI-associated lung permeability in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Human pulmonary microvascular endothelial cell (HPMVEC) monolayers were incubated with IMD, and transcellular electrical resistance was measured to quantify endothelial barrier function. Expression and localization of endogenous pulmonary IMD, and its receptor complexes composed of calcitonin receptor-like receptor (CRLR) and receptor activity-modifying proteins (RAMPs) 1-3 were analyzed by qRT-PCR and immunofluorescence in non ventilated mouse lungs and in lungs ventilated for 6 h. In untreated and IMD treated mice, lung permeability, pulmonary leukocyte recruitment and cytokine levels were assessed after mechanical ventilation. Further, the impact of IMD on pulmonary vasoconstriction was investigated in precision cut lung slices (PCLS) and in isolated perfused and ventilated mouse lungs. IMD stabilized endothelial barrier function in HPMVECs. Mechanical ventilation reduced the expression of RAMP3, but not of IMD, CRLR, and RAMP1 and 2. Mechanical ventilation induced lung hyperpermeability, which was ameliorated by IMD treatment. Oxygenation was not improved by IMD, which may be attributed to impaired hypoxic vasoconstriction due to IMD treatment. IMD had minor impact on pulmonary leukocyte recruitment and did not reduce cytokine levels in VILI. CONCLUSIONS/SIGNIFICANCE: IMD may possibly provide a new approach to attenuate VILI.


Subject(s)
Endothelial Cells/drug effects , Lung Injury/prevention & control , Lung/drug effects , Peptide Hormones/pharmacology , Animals , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Female , Fluorescent Antibody Technique , Gene Expression/drug effects , Humans , Hypoxia , In Vitro Techniques , Lung/blood supply , Lung/metabolism , Lung Injury/etiology , Lung Injury/genetics , Mice , Mice, Inbred C57BL , Microvessels/cytology , Microvessels/drug effects , Microvessels/physiopathology , Peptide Hormones/genetics , Peptide Hormones/metabolism , Receptor Activity-Modifying Protein 1/genetics , Receptor Activity-Modifying Protein 1/metabolism , Receptor Activity-Modifying Protein 2/genetics , Receptor Activity-Modifying Protein 2/metabolism , Receptor Activity-Modifying Protein 3/genetics , Receptor Activity-Modifying Protein 3/metabolism , Respiration, Artificial/adverse effects , Reverse Transcriptase Polymerase Chain Reaction , Vasoconstriction/drug effects
10.
Cell Microbiol ; 13(11): 1668-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21790939

ABSTRACT

Defence mechanisms against intracellular bacterial pathogens are incompletely understood. Our study characterizes a type I IFN-dependent cell-autonomous defence pathway directed against Legionella pneumophila, an intracellular model organism and frequent cause of pneumonia. We show that macrophages infected with L. pneumophila produced IFNß in a STING- and IRF3- dependent manner. Paracrine type I IFNs stimulated upregulation of IFN-stimulated genes and a cell-autonomous defence pathway acting on replicating and non-replicating Legionella within their specialized vacuole. Our infection experiments in mice lacking receptors for type I and/or II IFNs show that type I IFNs contribute to expression of IFN-stimulated genes and to bacterial clearance as well as resistance in L. pneumophila pneumonia in addition to type II IFN. Overall, our study shows that paracrine type I IFNs mediate defence against L. pneumophila, and demonstrates a protective role of type I IFNs in in vivo infections with intracellular bacteria.


Subject(s)
Interferon Type I/immunology , Legionella pneumophila/immunology , Legionella pneumophila/pathogenicity , Legionnaires' Disease/immunology , Legionnaires' Disease/pathology , Signal Transduction , Animals , Disease Models, Animal , Gene Expression Profiling , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Macrophages/microbiology , Membrane Proteins/metabolism , Mice , Vacuoles/immunology , Vacuoles/microbiology
11.
Pulm Pharmacol Ther ; 24(4): 377-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21419858

ABSTRACT

Lung barrier protection by Sphingosine-1 Phosphate (S1P) has been demonstrated experimentally, but recent evidence suggests barrier disruptive properties of high systemic S1P concentrations. The S1P analog FTY720 recently gained an FDA approval for treatment of multiple sclerosis. In case of FTY720 treated patients experiencing multiple organ dysfunction syndrome the drug may accumulate due to liver failure, and the patients may receive ventilator therapy. Whereas low doses of FTY720 enhanced endothelial barrier function, data on effects of increased FTY720 concentrations are lacking. We measured transcellular electrical resistance (TER) of human umbilical vein endothelial cell (HUVEC) monolayers, performed morphologic analysis and measured apoptosis by TUNEL staining and procaspase-3 degradation in HUVECs stimulated with FTY720 (0.01-100 µM). Healthy C57BL/6 mice and mice ventilated with 17 ml/kg tidal volume and 100% oxygen for 2 h were treated with 0.1 or 2 mg/kg FTY720 or solvent, and lung permeability, oxygenation and leukocyte counts in BAL and blood were quantified. Further, electron microscopic analysis of lung tissue was performed. We observed barrier protective effects of FTY720 on HUVEC cell layers at concentrations up to 1 µM while higher concentrations induced irreversible barrier breakdown accompanied by induction of apoptosis. Low FTY720 concentrations (0.1 mg/kg) reduced lung permeability in mechanically ventilated mice, but 2 mg/kg FTY720 increased pulmonary vascular permeability in ventilated mice accompanied by endothelial apoptosis, while not affecting permeability in non-ventilated mice. Moreover, hyperoxic mechanical ventilation sensitized the pulmonary vasculature to a barrier disrupting effect of FTY720, resulting in worsening of ventilator induced lung injury. In conclusion, the current data suggest FTY720 induced endothelial barrier dysfunction, which was probably caused by proapoptotic effects and enhanced by mechanical ventilation.


Subject(s)
Endothelial Cells/drug effects , Lung/drug effects , Propylene Glycols/toxicity , Receptors, Lysosphingolipid/agonists , Sphingosine/analogs & derivatives , Ventilator-Induced Lung Injury/etiology , Animals , Apoptosis/drug effects , Capillary Permeability/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Fingolimod Hydrochloride , Humans , Lung/pathology , Mice , Mice, Inbred C57BL , Sphingosine/toxicity
12.
Crit Care ; 14(4): R143, 2010.
Article in English | MEDLINE | ID: mdl-20673352

ABSTRACT

INTRODUCTION: Mechanical ventilation (MV) is a life saving intervention in acute respiratory failure without alternative. However, particularly in pre-injured lungs, even protective ventilation strategies may evoke ventilator-induced lung injury (VILI), which is characterized by pulmonary inflammation and vascular leakage. Adjuvant pharmacologic strategies in addition to lung protective ventilation to attenuate VILI are lacking. Simvastatin exhibited anti-inflammatory and endothelial barrier stabilizing properties in vitro and in vivo. METHODS: Mice were ventilated (12 ml/kg; six hours) and subjected to simvastatin (20 mg/kg) or sham treatment. Pulmonary microvascular leakage, oxygenation, pulmonary and systemic neutrophil and monocyte counts and cytokine release in lung and blood plasma were assessed. Further, lung tissue was analyzed by electron microscopy. RESULTS: Mechanical ventilation induced VILI, displayed by increased pulmonary microvascular leakage and endothelial injury, pulmonary recruitment of neutrophils and Gr-1high monocytes, and by liberation of inflammatory cytokines in the lungs. Further, VILI associated systemic inflammation characterized by blood leukocytosis and elevated plasma cytokines was observed. Simvastatin treatment limited pulmonary endothelial injury, attenuated pulmonary hyperpermeability, prevented the recruitment of leukocytes to the lung, reduced pulmonary cytokine levels and improved oxygenation in mechanically ventilated mice. CONCLUSIONS: High-dose simvastatin attenuated VILI in mice by reducing MV-induced pulmonary inflammation and hyperpermeability.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Simvastatin/therapeutic use , Ventilator-Induced Lung Injury/drug therapy , Alanine Transaminase/blood , Animals , Blood Gas Analysis , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cytokines/blood , Female , Leukocyte Count , Mice , Mice, Inbred C57BL , Microscopy, Electron , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Respiratory Mucosa/physiopathology , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...