Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nat Commun ; 15(1): 3450, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664395

ABSTRACT

Influenza A viruses (IAVs) of subtype H9N2 have reached an endemic stage in poultry farms in the Middle East and Asia. As a result, human infections with avian H9N2 viruses have been increasingly reported. In 2017, an H9N2 virus was isolated for the first time from Egyptian fruit bats (Rousettus aegyptiacus). Phylogenetic analyses revealed that bat H9N2 is descended from a common ancestor dating back centuries ago. However, the H9 and N2 sequences appear to be genetically similar to current avian IAVs, suggesting recent reassortment events. These observations raise the question of the zoonotic potential of the mammal-adapted bat H9N2. Here, we investigate the infection and transmission potential of bat H9N2 in vitro and in vivo, the ability to overcome the antiviral activity of the human MxA protein, and the presence of N2-specific cross-reactive antibodies in human sera. We show that bat H9N2 has high replication and transmission potential in ferrets, efficiently infects human lung explant cultures, and is able to evade antiviral inhibition by MxA in transgenic B6 mice. Together with its low antigenic similarity to the N2 of seasonal human strains, bat H9N2 fulfils key criteria for pre-pandemic IAVs.


Subject(s)
Chiroptera , Ferrets , Influenza A Virus, H9N2 Subtype , Orthomyxoviridae Infections , Virus Replication , Animals , Ferrets/virology , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/physiology , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/isolation & purification , Chiroptera/virology , Humans , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/immunology , Mice , Phylogeny , Influenza, Human/transmission , Influenza, Human/virology , Lung/virology , Antibodies, Viral/immunology , Antibodies, Viral/blood
2.
Sci Rep ; 12(1): 20608, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36446841

ABSTRACT

Influenza A virus (IAV) causes pandemics and annual epidemics of severe respiratory infections. A better understanding of the molecular regulation in tissue and cells upon IAV infection is needed to thoroughly understand pathogenesis. We analyzed IAV replication and gene expression induced by IAV strain H3N2 Panama in isolated primary human alveolar epithelial type II cells (AECIIs), the permanent A549 adenocarcinoma cell line, alveolar macrophages (AMs) and explanted human lung tissue by bulk RNA sequencing. Primary AECII exhibit in comparison to AM a broad set of strongly induced genes related to RIG-I and interferon (IFN) signaling. The response of AECII was partly mirrored in A549 cells. In human lung tissue, we observed induction of genes unlike in isolated cells. Viral RNA was used to correlate host cell gene expression changes with viral burden. While relative induction of key genes was similar, gene abundance was highest in AECII cells and AM, while weaker in the human lung (due to less IAV replication) and A549 cells (pointing to their limited suitability as a model). Correlation of host gene induction with viral burden allows a better understanding of the cell-type specific induction of pathways and a possible role of cellular crosstalk requiring intact tissue.


Subject(s)
Influenza A virus , Influenza, Human , Humans , A549 Cells , Transcriptome , Influenza A Virus, H3N2 Subtype , Alveolar Epithelial Cells , Influenza, Human/genetics
3.
Commun Biol ; 5(1): 875, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36008580

ABSTRACT

Mechanisms of epithelial renewal in the alveolar compartment remain incompletely understood. To this end, we aimed to characterize alveolar progenitors. Single-cell RNA-sequencing (scRNA-seq) analysis of the HTII-280+/EpCAM+ population from adult human lung revealed subclusters enriched for adult stem cell signature (ASCS) genes. We found that alveolar progenitors in organoid culture in vitro show phenotypic lineage plasticity as they can yield alveolar or bronchial cell-type progeny. The direction of the differentiation is dependent on the presence of the GSK-3ß inhibitor, CHIR99021. By RNA-seq profiling of GSK-3ß knockdown organoids we identified additional candidate target genes of the inhibitor, among others FOXM1 and EGF. This gives evidence of Wnt pathway independent regulatory mechanisms of alveolar specification. Following influenza A virus (IAV) infection organoids showed a similar response as lung tissue explants which confirms their suitability for studies of sequelae of pathogen-host interaction.


Subject(s)
Lung , Organoids , Cell Differentiation/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Lung/metabolism , Organoids/metabolism , Wnt Signaling Pathway
4.
Eur Respir Rev ; 31(165)2022 Sep 30.
Article in English | MEDLINE | ID: mdl-35896273

ABSTRACT

Single-cell ribonucleic acid sequencing is becoming widely employed to study biological processes at a novel resolution depth. The ability to analyse transcriptomes of multiple heterogeneous cell types in parallel is especially valuable for cell-focused lung research where a variety of resident and recruited cells are essential for maintaining organ functionality. We compared the single-cell transcriptomes from publicly available and unpublished datasets of the lungs in six different species: human (Homo sapiens), African green monkey (Chlorocebus sabaeus), pig (Sus domesticus), hamster (Mesocricetus auratus), rat (Rattus norvegicus) and mouse (Mus musculus) by employing RNA velocity and intercellular communication based on ligand-receptor co-expression, among other techniques. Specifically, we demonstrated a workflow for interspecies data integration, applied a single unified gene nomenclature, performed cell-specific clustering and identified marker genes for each species. Overall, integrative approaches combining newly sequenced as well as publicly available datasets could help identify species-specific transcriptomic signatures in both healthy and diseased lung tissue and select appropriate models for future respiratory research.


Subject(s)
Pulmonologists , Transcriptome , Animals , Base Sequence , Chlorocebus aethiops , Cricetinae , Humans , Lung , Mice , Rats , Species Specificity , Swine
5.
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
6.
Zentralbl Chir ; 145(1): 82-88, 2020 Feb.
Article in German | MEDLINE | ID: mdl-31269514

ABSTRACT

INTRODUCTION: Infectious complications after lung resections pose a high burden of perioperative morbidity and mortality. Among other factors, perioperative antibiotic prophylaxis and management of a postoperative pneumonia have an impact on patient outcome. We developed a local clinical pathway for adequate perioperative use of antibiotics. METHODS: We analysed respiratory samples of 200 patients taken before and after lung resection performed in our lung clinic from October 2013 till October 2014. The clinical pathway was based on our local pathogen and resistance pattern as well as on current guidelines and on the principals of antibiotic stewardship. RESULTS: Gram negative bacteria were the predominant pathogens that grew from the samples in the preoperative phase (62%), as well as in the postoperative phase (78%). A significant number of these bacteria showed intrinsic resistance against the commonly used antibiotics for perioperative prophylaxis. This was the case for both the preoperative phase (21%) and the postoperative phase (39%). These findings were integrated into the local clinical pathway. CONCLUSION: The commonly used antibiotics for perioperative prophylaxis in thoracic surgery cover only some of the pathogens responsible for preoperative airway colonisation and postoperative pneumonia. Therefore, perioperative antibiotic prophylaxis should be given as a single shot just before surgery and postoperative pneumonia should be treated as a hospital acquired pneumonia with respect to the local pathogen and resistance pattern.


Subject(s)
Thoracic Surgical Procedures , Anti-Bacterial Agents , Antibiotic Prophylaxis , Humans , Postoperative Complications , Prospective Studies , Thoracic Surgery
7.
Front Immunol ; 9: 1346, 2018.
Article in English | MEDLINE | ID: mdl-29977236

ABSTRACT

Early immune responses to Mycobacterium tuberculosis (Mtb) invasion of the human lung play a decisive role in the outcome of infection, leading to either rapid clearance of the pathogen or stable infection. Despite their critical impact on health and disease, these early host-pathogen interactions at the primary site of infection are still poorly understood. In vitro studies cannot fully reflect the complexity of the lung architecture and its impact on host-pathogen interactions, while animal models have their own limitations. In this study, we have investigated the initial responses in human lung tissue explants to Mtb infection, focusing primarily on gene expression patterns in different tissue-resident cell types. As first cell types confronted with pathogens invading the lung, alveolar macrophages, and epithelial cells displayed rapid proinflammatory chemokine and cytokine responses to Mtb infection. Other tissue-resident innate cells like gamma/delta T cells, mucosal associated invariant T cells, and natural killer cells showed partially similar but weaker responses, with a high degree of variability across different donors. Finally, we investigated the responses of tissue-resident innate lymphoid cells to the inflammatory milieu induced by Mtb infection. Our infection model provides a unique approach toward host-pathogen interactions at the natural port of Mtb entry and site of its implantation, i.e., the human lung. Our data provide a first detailed insight into the early responses of different relevant pulmonary cells in the alveolar microenvironment to contact with Mtb. These results can form the basis for the identification of host markers that orchestrate early host defense and provide resistance or susceptibility to stable Mtb infection.

8.
Emerg Microbes Infect ; 7(1): 132, 2018 Jul 19.
Article in English | MEDLINE | ID: mdl-30026505

ABSTRACT

We investigated in a unique setup of animal models and a human lung explant culture biological properties, including zoonotic potential, of a representative 2016 highly pathogenic avian influenza virus (HPAIV) H5N8, clade 2.3.4.4 group B (H5N8B), that spread rapidly in a huge and ongoing outbreak series in Europe and caused high mortality in waterfowl and domestic birds. HPAIV H5N8B showed increased virulence with rapid onset of severe disease and mortality in Pekin ducks due to pronounced neuro- and hepatotropism. Cross-species infection was evaluated in mice, ferrets, and in a human lung explant culture model. While the H5N8B isolate was highly virulent for Balb/c mice, virulence and transmissibility were grossly reduced in ferrets, which was mirrored by marginal replication in human lung cultures infected ex vivo. Our data indicate that the 2016 HPAIV H5N8B is avian-adapted with augmented virulence for waterfowl, but has low zoonotic potential. The here tested combination of animal studies with the inoculation of human explants provides a promising future workflow to evaluate zoonotic potential, mammalian replication competence and avian virulence of HPAIV.


Subject(s)
Ducks/virology , Influenza A Virus, H5N8 Subtype/pathogenicity , Influenza in Birds/transmission , Influenza, Human/transmission , Poultry Diseases/virology , Zoonoses/transmission , Zoonoses/virology , Animals , Disease Outbreaks/veterinary , Ferrets/virology , Humans , Influenza in Birds/virology , Influenza, Human/virology , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Poultry Diseases/transmission , Virulence , Virus Replication
9.
Sci Rep ; 8(1): 182, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29317705

ABSTRACT

Streptococcus pneumoniae (S.pn.) is the most common bacterial pathogen causing community acquired pneumonia. The pore-forming toxin pneumolysin (PLY) is the major virulence factor of S.pn. and supposed to affect alveolar epithelial cells thereby activating the immune system by liberation of danger-associated molecular patterns (DAMP). To test this hypothesis, we established a novel live-cell imaging based assay to analyse mitochondrial function and associated release of mitochondrial DNA (mtDNA) as DAMP in real-time. We first revealed that bacterially released PLY caused significant changes of the cellular ATP homeostasis and led to morphologic alterations of mitochondria in human alveolar epithelial cells in vitro and, by use of spectral live-tissue imaging, in human alveoli. This was accompanied by strong mitochondrial calcium influx and loss of mitochondrial membrane potential resulting in opening of the mitochondrial permeability transition pore and mtDNA release without activation of intrinsic apoptosis. Moreover, our data indicate cellular mtDNA liberation via microvesicles, which may contribute to S.pn. related pro-inflammatory immune activation in the human alveolar compartment.


Subject(s)
Alveolar Epithelial Cells/drug effects , DNA, Mitochondrial/metabolism , Mitochondria/drug effects , Streptolysins/toxicity , Adenosine Triphosphate/metabolism , Alveolar Epithelial Cells/metabolism , Bacterial Proteins/toxicity , Calcium/metabolism , Cell Line, Tumor , Cells, Cultured , Humans , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore
10.
Crit Care Med ; 46(3): e258-e267, 2018 03.
Article in English | MEDLINE | ID: mdl-29298188

ABSTRACT

OBJECTIVES: Severe pneumonia may evoke acute lung injury, and sphingosine-1-phosphate is involved in the regulation of vascular permeability and immune responses. However, the role of sphingosine-1-phosphate and the sphingosine-1-phosphate producing sphingosine kinase 1 in pneumonia remains elusive. We examined the role of the sphingosine-1-phosphate system in regulating pulmonary vascular barrier function in bacterial pneumonia. DESIGN: Controlled, in vitro, ex vivo, and in vivo laboratory study. SUBJECTS: Female wild-type and SphK1-deficient mice, 8-10 weeks old. Human postmortem lung tissue, human blood-derived macrophages, and pulmonary microvascular endothelial cells. INTERVENTIONS: Wild-type and SphK1-deficient mice were infected with Streptococcus pneumoniae. Pulmonary sphingosine-1-phosphate levels, messenger RNA expression, and permeability as well as lung morphology were analyzed. Human blood-derived macrophages and human pulmonary microvascular endothelial cells were infected with S. pneumoniae. Transcellular electrical resistance of human pulmonary microvascular endothelial cell monolayers was examined. Further, permeability of murine isolated perfused lungs was determined following exposition to sphingosine-1-phosphate and pneumolysin. MEASUREMENTS AND MAIN RESULTS: Following S. pneumoniae infection, murine pulmonary sphingosine-1-phosphate levels and sphingosine kinase 1 and sphingosine-1-phosphate receptor 2 expression were increased. Pneumonia-induced lung hyperpermeability was reduced in SphK1 mice compared with wild-type mice. Expression of sphingosine kinase 1 in macrophages recruited to inflamed lung areas in pneumonia was observed in murine and human lungs. S. pneumoniae induced the sphingosine kinase 1/sphingosine-1-phosphate system in blood-derived macrophages and enhanced sphingosine-1-phosphate receptor 2 expression in human pulmonary microvascular endothelial cell in vitro. In isolated mouse lungs, pneumolysin-induced hyperpermeability was dose dependently and synergistically increased by sphingosine-1-phosphate. This sphingosine-1-phosphate-induced increase was reduced by inhibition of sphingosine-1-phosphate receptor 2 or its downstream effector Rho-kinase. CONCLUSIONS: Our data suggest that targeting the sphingosine kinase 1-/sphingosine-1-phosphate-/sphingosine-1-phosphate receptor 2-signaling pathway in the lung may provide a novel therapeutic perspective in pneumococcal pneumonia for prevention of acute lung injury.


Subject(s)
Acute Lung Injury/metabolism , Inflammation/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pneumonia, Pneumococcal/metabolism , Receptors, Lysosphingolipid/metabolism , Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Animals , Female , Humans , Inflammation/enzymology , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/enzymology , Sphingosine-1-Phosphate Receptors , Streptococcus pneumoniae
11.
Eur Respir J ; 50(1)2017 07.
Article in English | MEDLINE | ID: mdl-28705941

ABSTRACT

The severity and lethality of influenza A virus (IAV) infections is frequently aggravated by secondary bacterial pneumonia. However, the mechanisms in human lung tissue that provoke this increase in fatality are unknown and therapeutic immune modulatory options are lacking.We established a human lung ex vivo co-infection model to investigate innate immune related mechanisms contributing to the susceptibility of secondary pneumococcal pneumonia.We revealed that type I and III interferon (IFN) inhibits Streptococcus pneumoniae-induced interleukin (IL)-1ß release. The lack of IL-1ß resulted in the repression of bacterially induced granulocyte-macrophage colony-stimulating factor (GM-CSF) liberation. Specific inhibition of IFN receptor I and III-associated tyrosine kinase 2 (Tyk2) completely restored the S. pneumoniae-induced IL-1ß-GM-CSF axis, leading to a reduction of bacterial growth. A preceding IAV infection of the human alveolus leads to a type I and III IFN-dependent blockade of the early cytokines IL-1ß and GM-CSF, which are key for orchestrating an adequate innate immune response against bacteria. Their virally induced suppression may result in impaired bacterial clearance and alveolar repair.Pharmacological inhibition of Tyk2 might be a new treatment option to sustain beneficial endogenous GM-CSF levels in IAV-associated secondary bacterial pneumonia.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Influenza, Human/drug therapy , Interferons/pharmacology , Pneumonia, Bacterial/drug therapy , Staphylococcal Infections/drug therapy , TYK2 Kinase/antagonists & inhibitors , Humans , Immunity, Innate/drug effects , Immunologic Factors , Influenza A virus , Influenza, Human/immunology , Interleukin-1beta/metabolism , Lung/drug effects , Pneumonia, Bacterial/immunology , Staphylococcal Infections/immunology , TYK2 Kinase/metabolism
12.
Lung Cancer ; 93: 28-34, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26898611

ABSTRACT

OBJECTIVES: Surgery has been available for the treatment of mono-metastatic, non-small cell lung cancer (NSCLC) and promising overall survival was observed in some retrospective studies with selected patients. This study investigated whether the preoperative 18-fluorodeoxyglucose positron emission tomography/computed tomography ((18)F-FDG-PET/CT) scan influences survival in this patient group. Furthermore we tried to identify other prognostic factors associated with survival and aimed to clarify if synchronous metastases are different from metachronous disease. METHODS: Between 1994 and 2012, 181 patients underwent resection for solitary metastases. Sixty-six patients underwent surgery after an initial FDG-PET/CT scan, whereas 115 patients underwent conventional preoperative staging by a spiral CT scan. RESULTS: The overall 5-year survival rate was 38.8%. The 5-year survival rates after preoperative evaluation by FDG-PET/CT and by conventional CT were 58% and 33%, respectively (p=0.01). A higher 5-year survival rate was observed in patients without thoracic lymph node involvement (pN0: 44% vs. pN1-3: 33%, p=0.028). In patients with a solitary pulmonary metastasis, we observed a 5-year survival rate of 45.7%, whereas in patients with extrapulmonary metastases, the 5-year survival rate was 27.1% (p=0.001). In patients with a locally limited primary lung cancer according to the pT descriptor, we observed a 5-year survival rate of 53.1%, whereas in patients with a pT>1 descriptor, the 5-year survival rate was 33.6% (p=0.016). By multivariate analyses, we showed that preoperative FDG-PET/CT evaluation, no thoracic lymph node metastases, and sole pulmonary metastatic disease were favorable predictors of survival, whereas the time of metastasis (synchronous vs. metachronous) and maximum standardized uptake value was not. CONCLUSIONS: We conclude that resection of the primary tumor and metastasectomy for mono-metastatic NSCLC can be performed after a comprehensive evaluation with FDG-PET/CT. N-stage and the site of the oligometastases have a significant influence on overall survival.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/mortality , Fluorodeoxyglucose F18 , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Positron-Emission Tomography , Tomography, X-Ray Computed , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Non-Small-Cell Lung/surgery , Female , Humans , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Male , Middle Aged , Preoperative Care , Prognosis , Survival Analysis , Tumor Burden
13.
Thorac Cardiovasc Surg Rep ; 4(1): 5-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26693117

ABSTRACT

Pneumocephalus can be seen after head injury with fracture of the skull-base or in cerebral neoplasm, infection, or after intracranial or spinal surgery. We report on a 69-year-old male patient with pneumocephalus after right-sided lobectomy and en bloc resection of the chest wall for non-small-cell lung cancer. Postoperatively, the patient showed a reduced vigilance level with no response to pain stimuli and anisocoria. The CCT scan revealed an extensive pneumocephalus; following which, the patient underwent neurosurgery with laminectomy and ligature of the transected nerve roots. After operation the patient returned to his baseline mental status.

15.
Ann Thorac Surg ; 98(1): 249-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24820385

ABSTRACT

BACKGROUND: Surgical treatment of patients with limited metastatic lesions from non-small cell lung cancer (NSCLC) remains controversial; however, reports suggest that a subset of patients may benefit from complete resection including metastasectomy. METHODS: Between 1997 and 2009, 99 patients underwent complete solitary synchronous NSCLC metastasis resection in a single center. Only patients who met the potentially curative operation criteria (ie, primary NSCLC and metastasis resection of a solitary pulmonary or solitary extrapulmonary metastases) were included for retrospective analyses within this study. RESULTS: The overall 5-year survival rate was 38%. A significantly longer survival was observed in patients without mediastinal (N2 or N3) lymph node involvement (median, 50.0 months) compared with patients who had mediastinal lymph node metastases (median, 19.0 months survival; p=0.015). In patients with a solitary metastasis in the ipsilateral (not ipsilobar) or contralateral lung, we observed a 5-year survival rate of 48.5%, whereas the rate was 23.6% in patients with extrapulmonary metastases (p=0.006). In univariate analysis, a trend for a more favorable long-term survival rate was observed for patients with a histologic grade of G1 or G2 versus G3 primary NSCLC (p=0.058). CONCLUSIONS: We conclude that metastasectomy for synchronous oligometastatic disease in NSCLC can be performed in selected patients. It appears reasonable that such patients should be considered as surgical candidates if mediastinal lymph node involvement is excluded.


Subject(s)
Carcinoma, Non-Small-Cell Lung/surgery , Lung Neoplasms/surgery , Metastasectomy/methods , Neoplasms, Multiple Primary/surgery , Pneumonectomy/methods , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Female , Follow-Up Studies , Germany/epidemiology , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Mediastinum , Middle Aged , Neoplasms, Multiple Primary/mortality , Retrospective Studies , Survival Rate/trends , Time Factors , Treatment Outcome
17.
mBio ; 4(5): e00601-13, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24105764

ABSTRACT

UNLABELLED: A novel influenza A virus (IAV) of the H7N9 subtype has been isolated from severely diseased patients with pneumonia and acute respiratory distress syndrome and, apparently, from healthy poultry in March 2013 in Eastern China. We evaluated replication, tropism, and cytokine induction of the A/Anhui/1/2013 (H7N9) virus isolated from a fatal human infection and two low-pathogenic avian H7 subtype viruses in a human lung organ culture system mimicking infection of the lower respiratory tract. The A(H7N9) patient isolate replicated similarly well as a seasonal IAV in explanted human lung tissue, whereas avian H7 subtype viruses propagated poorly. Interestingly, the avian H7 strains provoked a strong antiviral type I interferon (IFN-I) response, whereas the A(H7N9) virus induced only low IFN levels. Nevertheless, all viruses analyzed were detected predominantly in type II pneumocytes, indicating that the A(H7N9) virus does not differ in its cellular tropism from other avian or human influenza viruses. Tissue culture-based studies suggested that the low induction of the IFN-ß promoter correlated with an efficient suppression by the viral NS1 protein. These findings demonstrate that the zoonotic A(H7N9) virus is unusually well adapted to efficient propagation in human alveolar tissue, which most likely contributes to the severity of lower respiratory tract disease seen in many patients. IMPORTANCE: Humans are usually not infected by avian influenza A viruses (IAV), but this large group of viruses contributes to the emergence of human pandemic strains. Transmission of virulent avian IAV to humans is therefore an alarming event that requires assessment of the biology as well as pathogenic and pandemic potentials of the viruses in clinically relevant models. Here, we demonstrate that an early virus isolate from the recent A(H7N9) outbreak in Eastern China replicated as efficiently as human-adapted IAV in explanted human lung tissue, whereas avian H7 subtype viruses were unable to propagate. Robust replication of the H7N9 strain correlated with a low induction of antiviral beta interferon (IFN-ß), and cell-based studies indicated that this is due to efficient suppression of the IFN response by the viral NS1 protein. Thus, explanted human lung tissue appears to be a useful experimental model to explore the determinants facilitating cross-species transmission of the H7N9 virus to humans.


Subject(s)
Influenza A virus/growth & development , Influenza, Human/virology , Lung/virology , Animals , Birds , Cell Line , China , Humans , Influenza A virus/isolation & purification , Influenza A virus/physiology , Influenza in Birds/virology , Influenza, Human/immunology , Influenza, Human/pathology , Interferon-beta/immunology , Lung/immunology , Lung/pathology , Molecular Sequence Data , Virus Replication
19.
J Infect Dis ; 206(11): 1685-94, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22829640

ABSTRACT

BACKGROUND: Highly pathogenic avian H5N1 influenza viruses preferentially infect alveolar type II pneumocytes in human lung. However, it is unknown whether this cellular tropism contributes to high viral virulence because the primary target cells of other influenza viruses have not been systematically studied. METHODS: We provide the first comparison of the replication, tropism, and cytokine induction of human, highly pathogenic avian influenza A virus subtype H5N1 and other animal influenza A viruses in primary human lung organ cultures. RESULTS: Subytpe H5N1 and human-adapted subtype H1N1 and H3N2 viruses replicated efficiently in the lung tissue, whereas classic swine and low-pathogenicity avian viruses propagated only poorly. Nevertheless, all viruses examined were detected almost exclusively in type II pneumocytes, with a minor involvement of alveolar macrophages. Infection with avian viruses that have a low and high pathogenicity provoked a pronounced induction of cytokines and chemokines, while human and pandemic H1N1-2009 viruses triggered only weak responses. CONCLUSIONS: These findings show that differences in the pathogenic potential of influenza A viruses in the human lung cannot be attributed to a distinct cellular tropism. Rather, high or low viral pathogenicity is associated with a strain-specific capacity to productively replicate in type II pneumocytes and to cope with the induced cytokine response.


Subject(s)
Alveolar Epithelial Cells/classification , Alveolar Epithelial Cells/virology , Influenza A virus/physiology , Viral Tropism/physiology , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/physiology , Humans , Influenza A virus/classification , Influenza A virus/pathogenicity , Influenza, Human/virology , Lung/cytology , Macrophages, Alveolar/virology , Tissue Culture Techniques , Virulence , Virus Replication/physiology
20.
Toxicol Lett ; 164(3): 278-88, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16483733

ABSTRACT

The lung represents an important target for the toxic effects of chemicals. Many of the chemicals require enzymatic activation to exert their adverse effects, which is mostly catalysed by Cytochrome P450 (CYP) enzymes. Although there is considerable evidence that individual members of the xenobiotic-metabolizing P450 family are expressed in human lung tissue at the mRNA level, there is conflicting evidence concerning the following issues: (I) the qualitative expression pattern of CYP isoenzymes; (II) CYP expression at the protein and/or activity level; and (III) interindividual variability of CYP enzymes in human lung. The latter can be the basis for individual susceptibility towards the adverse effects of lung toxicants. In preparing for studying factors to explain interindividual variability of CYP expression in lung tissue, we investigated the qualitative pulmonary expression pattern of xenobiotic-metabolizing CYP enzymes and elaborated the optimal conditions for quantification at the protein and activity level. By using either individual human lung samples or pooled microsomes from different individuals, immunoreactive bands specific for the following CYP enzymes could be determined by Western blotting: CYP1A1, CYP1A2, CYP2E1 and CYP3A5. Western blotting experiments were also supportive of the presence of CYP2A, CYP2B6, CYP2D6 and CYP3A4 in human lung. By using antibodies specific for CYP2C enzymes and CYP1B1, respectively, immunoreactive bands, which differed slightly in mobility from corresponding standards, were detectable. In addition, we measured methoxy- and ethoxyresorufin dealkylase activities and chlorzoxazone (CLX)-hydroxylase activity in human lung and confirmed the specifities of the latter two activities by inhibition experiments. In summary, we have established methodologies to quantify a panel of CYP enzymes in human lung samples among which there are CYP enzymes whose expression at the protein and activity level has not been evidenced so far.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Lung/drug effects , Xenobiotics/pharmacokinetics , Blotting, Western , Cytochrome P-450 Enzyme System/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Inactivation, Metabolic , Lung/cytology , Lung/enzymology , Microsomes/drug effects , Microsomes/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...