Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cardiovasc Res ; 2022 Aug 23.
Article in English | MEDLINE | ID: covidwho-2256625

ABSTRACT

AIMS: SARS-CoV-2 infection causes COVID-19, which in severe cases evokes life-threatening acute respiratory distress syndrome (ARDS). Transcriptome signatures and the functional relevance of non-vascular cell types (e.g. immune and epithelial cells) in COVID-19 are becoming increasingly evident. However, despite its known contribution to vascular inflammation, recruitment/invasion of immune cells, vascular leakage and perturbed hemostasis in the lungs of severe COVID-19 patients, an in-depth interrogation of the endothelial cell (EC) compartment in lethal COVID-19 is lacking. Moreover, progressive fibrotic lung disease represents one of the complications of COVID-19 pneumonia and ARDS. Analogous features between idiopathic pulmonary fibrosis (IPF) and COVID-19 suggest partial similarities in their pathophysiology, yet, a head-to-head comparison of pulmonary cell transcriptomes between both conditions has not been implemented to date. METHODS AND RESULTS: We performed single nucleus RNA-seq (snRNA-seq) on frozen lungs from 7 deceased COVID-19 patients, 6 IPF explant lungs and 12 controls. The vascular fraction, comprising 38,794 nuclei, could be subclustered into 14 distinct EC subtypes. Non-vascular cell types, comprising 137,746 nuclei, were subclustered and used for EC-interactome analyses. Pulmonary ECs of deceased COVID-19 patients showed an enrichment of genes involved in cellular stress, as well as signatures suggestive of dampened immunomodulation and impaired vessel wall integrity. In addition, increased abundance of a population of systemic capillary and venous ECs was identified in COVID-19 and IPF. COVID-19 systemic ECs closely resembled their IPF counterparts, and a set of 30 genes was found congruently enriched in systemic ECs across studies. Receptor-ligand interaction analysis of ECs with non-vascular cell types in the pulmonary micro-environment revealed numerous previously unknown interactions specifically enriched/depleted in COVID-19 and/or IPF. CONCLUSIONS: This study uncovered novel insights into the abundance, expression patterns and interactomes of EC subtypes in COVID-19 and IPF, relevant for future investigations into the progression and treatment of both lethal conditions. TRANSLATIONAL PERSPECTIVE: While assessing clinical and molecular characteristics of severe and lethal COVID-19 cases, the vasculature's undeniable role in disease progression has been widely acknowledged. COVID-19 lung pathology moreover shares certain clinical features with late-stage IPF - yet an in-depth interrogation and direct comparison of the endothelium at single-cell level in both conditions is still lacking. By comparing the transcriptomes of ECs from lungs of deceased COVID-19 patients to those from IPF explant and control lungs, we gathered key insights the heterogeneous composition and potential roles of ECs in both lethal diseases, which may serve as a foundation for development of novel therapeutics.

2.
Front Immunol ; 12: 763098, 2021.
Article in English | MEDLINE | ID: covidwho-1581339

ABSTRACT

Although initial immunophenotypical studies on peripheral blood and bronchoalveolar lavage samples have provided a glimpse into the immunopathology of COVID-19, analyses of pulmonary draining lymph nodes are currently scarce. 22 lethal COVID-19 cases and 28 controls were enrolled in this study. Pulmonary draining lymph nodes (mediastinal, tracheal, peribronchial) were collected at autopsy. Control lymph nodes were selected from a range of histomorphological sequelae [unremarkable histology, infectious mononucleosis, follicular hyperplasia, non-SARS related HLH, extrafollicular plasmablast activation, non-SARS related diffuse alveolar damage (DAD), pneumonia]. Samples were mounted on a tissue microarray and underwent immunohistochemical staining for a selection of immunological markers and in-situ hybridization for Epstein Barr Virus (EBV) and SARS-CoV-2. Gene expression profiling was performed using the HTG EdgeSeq Immune Response Panel. Characteristic patterns of a dysregulated immune response were detected in COVID-19: 1. An accumulation of extrafollicular plasmablasts with a relative paucity or depletion of germinal centers. 2. Evidence of T-cell dysregulation demonstrated by immunohistochemical paucity of FOXP3+, Tbet+ and LEF1+ positive T-cells and a downregulation of key genes responsible for T-cell crosstalk, maturation and migration as well as a reactivation of herpes viruses in 6 COVID-19 lymph nodes (EBV, HSV). 3. Macrophage activation by a M2-polarized, CD163+ phenotype and increased incidence of hemophagocytic activity. 4. Microvascular dysfunction, evidenced by an upregulation of hemostatic (CD36, PROCR, VWF) and proangiogenic (FLT1, TEK) genes and an increase of fibrin microthrombi and CD105+ microvessels. Taken together, these findings imply widespread dysregulation of both innate and adoptive pathways with concordant microvascular dysfunction in severe COVID-19.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Lung , Macrophage Activation/immunology , Male , Middle Aged , SARS-CoV-2 , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thromboinflammation/immunology , Thromboinflammation/pathology , Thromboinflammation/virology
3.
Pathologe ; 43(2): 135-139, 2022 Mar.
Article in German | MEDLINE | ID: covidwho-1588808

ABSTRACT

We report a case of a placenta with extensive maternal vascular malperfusion and chronic histiocytic intervillositis corresponding to SARS-CoV­2 placentitis in the context of fetal demise at 31 weeks of gestation. Placental swamp and PCR of the placental parenchyma, umbilical cord and amnion-chorion membrane showed SARS-CoV-2- and B­betacoronavirus-specific RNA. Maternal vascular malperfusion has been described in cases of SARS-CoV­2 infection; however, the manifested severity of this case in the setting of a severe SARS-CoV­2 placentitis is rare. It emphasizes the need of a maternal prophylactic anticoagulation.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Female , Fetal Death , Humans , Placenta , Pregnancy , SARS-CoV-2 , Stillbirth
4.
EMBO Mol Med ; 13(11): e13714, 2021 11 08.
Article in English | MEDLINE | ID: covidwho-1471196

ABSTRACT

Risk stratification of COVID-19 patients is essential for pandemic management. Changes in the cell fitness marker, hFwe-Lose, can precede the host immune response to infection, potentially making such a biomarker an earlier triage tool. Here, we evaluate whether hFwe-Lose gene expression can outperform conventional methods in predicting outcomes (e.g., death and hospitalization) in COVID-19 patients. We performed a post-mortem examination of infected lung tissue in deceased COVID-19 patients to determine hFwe-Lose's biological role in acute lung injury. We then performed an observational study (n = 283) to evaluate whether hFwe-Lose expression (in nasopharyngeal samples) could accurately predict hospitalization or death in COVID-19 patients. In COVID-19 patients with acute lung injury, hFwe-Lose is highly expressed in the lower respiratory tract and is co-localized to areas of cell death. In patients presenting in the early phase of COVID-19 illness, hFwe-Lose expression accurately predicts subsequent hospitalization or death with positive predictive values of 87.8-100% and a negative predictive value of 64.1-93.2%. hFwe-Lose outperforms conventional inflammatory biomarkers and patient age and comorbidities, with an area under the receiver operating characteristic curve (AUROC) 0.93-0.97 in predicting hospitalization/death. Specifically, this is significantly higher than the prognostic value of combining biomarkers (serum ferritin, D-dimer, C-reactive protein, and neutrophil-lymphocyte ratio), patient age and comorbidities (AUROC of 0.67-0.92). The cell fitness marker, hFwe-Lose, accurately predicts outcomes in COVID-19 patients. This finding demonstrates how tissue fitness pathways dictate the response to infection and disease and their utility in managing the current COVID-19 pandemic.


Subject(s)
COVID-19 , Biomarkers , Flowers , Humans , Pandemics , ROC Curve , Retrospective Studies , SARS-CoV-2 , Severity of Illness Index
5.
Cell Rep Med ; 2(10): 100421, 2021 10 19.
Article in English | MEDLINE | ID: covidwho-1440413

ABSTRACT

Understanding viral tropism is an essential step toward reducing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission, decreasing mortality from coronavirus disease 2019 (COVID-19) and limiting opportunities for mutant strains to arise. Currently, little is known about the extent to which distinct tissue sites in the human head and neck region and proximal respiratory tract selectively permit SARS-CoV-2 infection and replication. In this translational study, we discover key variabilities in expression of angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2), essential SARS-CoV-2 entry factors, among the mucosal tissues of the human proximal airways. We show that SARS-CoV-2 infection is present in all examined head and neck tissues, with a notable tropism for the nasal cavity and tracheal mucosa. Finally, we uncover an association between smoking and higher SARS-CoV-2 viral infection in the human proximal airway, which may explain the increased susceptibility of smokers to developing severe COVID-19. This is at least partially explained by differences in interferon (IFN)-ß1 levels between smokers and non-smokers.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/transmission , Respiratory Mucosa/metabolism , Serine Endopeptidases/genetics , Smokers , Viral Tropism , Aged , Aged, 80 and over , COVID-19/genetics , COVID-19/metabolism , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Nasal Cavity/metabolism , SARS-CoV-2/physiology , Trachea/metabolism
6.
Front Cardiovasc Med ; 8: 695010, 2021.
Article in English | MEDLINE | ID: covidwho-1359167

ABSTRACT

We report on an impressive case of a previously healthy 47-year-old female Caucasian SARS-CoV-2 positive patient who died within 48 h after initial cardiac symptoms. Autopsy revealed necrotizing myocarditis and extensive microthrombosis as the cause of death. The interesting feature of this case is the combination of both myocarditis and extensive localized microthrombosis of cardiac capillaries. Microthrombosis was not present in other organs, and the patient did not show typical features of diffuse alveolar damage in the lungs. Taken together, our morphologic findings illustrate the angiocentric, microangiopathic, thromboinflammatory disease with significant thrombotic diathesis prevalent in COVID-19, which has been previously described in the literature, likely warranting thromboprophylaxis even in oligosymptomatic circumstances. This case also delineates several potential etiologies for microthrombosis, i.e., inflammatory reactions and primary hypercoagulative states. Further systematic analyses on risk stratification for receipt of prophylactic anticoagulation in COVID-19 are urgently required.

7.
Pathologe ; 42(2): 197-207, 2021 Mar.
Article in German | MEDLINE | ID: covidwho-1235733

ABSTRACT

BACKGROUND: The COVID-19 pandemic represents a so far unknown challenge for the medical community. Autopsies are important for studying this disease, but their safety was challenged at the beginning of the pandemic. OBJECTIVES: To determine whether COVID-19 autopsies can be performed under existing legal conditions and which safety standards are required. MATERIALS AND METHODS: The autopsy procedure undertaken in five institutions in Germany, Austria, and Switzerland is detailed with respect to legal and safety standards. RESULTS: In all institutions the autopsies were performed in technically feasible rooms. The personal equipment consisted of functional clothing including a disposable gown and apron, a surgical cap, eye protection, FFP­3 masks, and two pairs of gloves. In four institutions, complete autopsies were performed; in one institution the ultrasound-guided biopsy within the postmortal imaging and biopsy program. The latter does not allow the appreciation of gross organ pathology; however, it is able to retrieve standardized biopsies for diagnostic and research purposes. Several scientific articles in highly ranked journals resulted from these autopsies and allowed deep insights into organ damage and conclusions to better understand the pathomechanisms. Viral RNA was frequently detectable in the COVID-19 deceased, but the issue of infectivity remains unresolved and it is questionable if Ct values are greater than 30. CONCLUSIONS: With appropriate safeguards, autopsies of people who have died from COVID-19 can be performed safely and are highly relevant to medical research.


Subject(s)
COVID-19 , Pandemics , Austria , Autopsy , Germany , Humans , SARS-CoV-2 , Switzerland
8.
Radiol Cardiothorac Imaging ; 2(6): e200406, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-1156006

ABSTRACT

PURPOSE: The purpose of this retrospective study was to correlate CT patterns of fatal cases of coronavirus disease 2019 (COVID-19) with postmortem pathology observations. MATERIALS AND METHODS: The study included 70 lung lobes of 14 patients who died of reverse-transcription polymerase chain reaction-confirmed COVID-19. All patients underwent antemortem CT and autopsy between March 9 and April 30, 2020. Board-certified radiologists and pathologists performed lobewise correlations of pulmonary observations. In a consensus reading, 267 radiologic and 257 histopathologic observations of the lungs were recorded and systematically graded according to severity. These observations were matched and evaluated. RESULTS: Predominant CT observations were ground-glass opacities (GGO) (59/70 lobes examined) and areas of consolidation (33/70). The histopathologic observations were consistent with diffuse alveolar damage (70/70) and capillary dilatation and congestion (70/70), often accompanied by microthrombi (27/70), superimposed acute bronchopneumonia (17/70), and leukocytoclastic vasculitis (7/70). Four patients had pulmonary emboli. Bronchial wall thickening at CT histologically corresponded with acute bronchopneumonia. GGOs and consolidations corresponded with mixed histopathologic observations, including capillary dilatation and congestion, interstitial edema, diffuse alveolar damage, and microthrombosis. Vascular alterations were prominent observations at both CT and histopathology. CONCLUSION: A significant proportion of GGO correlated with the pathologic processes of diffuse alveolar damage, capillary dilatation and congestion, and microthrombosis. Our results confirm the presence and underline the importance of vascular alterations as key pathophysiologic drivers in lethal COVID-19.Supplemental material is available for this article.© RSNA, 2020.

10.
Histopathology ; 77(2): 198-209, 2020 Aug.
Article in English | MEDLINE | ID: covidwho-889739

ABSTRACT

AIMS: Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly evolved into a sweeping pandemic. Its major manifestation is in the respiratory tract, and the general extent of organ involvement and the microscopic changes in the lungs remain insufficiently characterised. Autopsies are essential to elucidate COVID-19-associated organ alterations. METHODS AND RESULTS: This article reports the autopsy findings of 21 COVID-19 patients hospitalised at the University Hospital Basel and at the Cantonal Hospital Baselland, Switzerland. An in-corpore technique was performed to ensure optimal staff safety. The primary cause of death was respiratory failure with exudative diffuse alveolar damage and massive capillary congestion, often accompanied by microthrombi despite anticoagulation. Ten cases showed superimposed bronchopneumonia. Further findings included pulmonary embolism (n = 4), alveolar haemorrhage (n = 3), and vasculitis (n = 1). Pathologies in other organ systems were predominantly attributable to shock; three patients showed signs of generalised and five of pulmonary thrombotic microangiopathy. Six patients were diagnosed with senile cardiac amyloidosis upon autopsy. Most patients suffered from one or more comorbidities (hypertension, obesity, cardiovascular diseases, and diabetes mellitus). Additionally, there was an overall predominance of males and individuals with blood group A (81% and 65%, respectively). All relevant histological slides are linked as open-source scans in supplementary files. CONCLUSIONS: This study provides an overview of postmortem findings in COVID-19 cases, implying that hypertensive, elderly, obese, male individuals with severe cardiovascular comorbidities as well as those with blood group A may have a lower threshold of tolerance for COVID-19. This provides a pathophysiological explanation for higher mortality rates among these patients.


Subject(s)
COVID-19/pathology , Capillaries/pathology , Vascular Diseases/pathology , Vascular Diseases/virology , Aged , Aged, 80 and over , Autopsy , Capillaries/virology , Female , Humans , Lung/pathology , Male , Middle Aged , SARS-CoV-2
11.
Nat Commun ; 11(1): 5086, 2020 10 08.
Article in English | MEDLINE | ID: covidwho-841091

ABSTRACT

Coronavirus Disease 19 (COVID-19) is a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has grown to a worldwide pandemic with substantial mortality. Immune mediated damage has been proposed as a pathogenic factor, but immune responses in lungs of COVID-19 patients remain poorly characterized. Here we show transcriptomic, histologic and cellular profiles of post mortem COVID-19 (n = 34 tissues from 16 patients) and normal lung tissues (n = 9 tissues from 6 patients). Two distinct immunopathological reaction patterns of lethal COVID-19 are identified. One pattern shows high local expression of interferon stimulated genes (ISGhigh) and cytokines, high viral loads and limited pulmonary damage, the other pattern shows severely damaged lungs, low ISGs (ISGlow), low viral loads and abundant infiltrating activated CD8+ T cells and macrophages. ISGhigh patients die significantly earlier after hospitalization than ISGlow patients. Our study may point to distinct stages of progression of COVID-19 lung disease and highlights the need for peripheral blood biomarkers that inform about patient lung status and guide treatment.


Subject(s)
Coronavirus Infections/immunology , Coronavirus Infections/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Aged , Aged, 80 and over , Betacoronavirus/pathogenicity , Betacoronavirus/physiology , CD8-Positive T-Lymphocytes/immunology , COVID-19 , Coronavirus Infections/mortality , Coronavirus Infections/virology , Cytokines/metabolism , Female , Gene Expression Profiling , Humans , Interferons/metabolism , Lung/immunology , Lung/pathology , Lung/virology , Macrophages/immunology , Male , Middle Aged , Pandemics , Pneumonia, Viral/mortality , Pneumonia, Viral/virology , SARS-CoV-2 , Viral Load
12.
Histopathology ; 78(3): 358-370, 2021 Feb.
Article in English | MEDLINE | ID: covidwho-796093

ABSTRACT

Transmission electron microscopy has become a valuable tool to investigate tissues of COVID-19 patients because it allows visualisation of SARS-CoV-2, but the 'virus-like particles' described in several organs have been highly contested. Because most electron microscopists in pathology are not accustomed to analysing viral particles and subcellular structures, our review aims to discuss the ultrastructural changes associated with SARS-CoV-2 infection and COVID-19 with respect to pathology, virology and electron microscopy. Using micrographs from infected cell cultures and autopsy tissues, we show how coronavirus replication affects ultrastructure and put the morphological findings in the context of viral replication, which induces extensive remodelling of the intracellular membrane systems. Virions assemble by budding into the endoplasmic reticulum-Golgi intermediate complex and are characterised by electron-dense dots of cross-sections of the nucleocapsid inside the viral particles. Physiological mimickers such as multivesicular bodies or coated vesicles serve as perfect decoys. Compared to other in-situ techniques, transmission electron microscopy is the only method to visualise assembled virions in tissues, and will be required to prove SARS-CoV-2 replication outside the respiratory tract. In practice, documenting in tissues the characteristic features seen in infected cell cultures seems to be much more difficult than anticipated. In our view, the hunt for coronavirus by transmission electron microscopy is still on.


Subject(s)
COVID-19/pathology , SARS-CoV-2/ultrastructure , COVID-19/virology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Humans , Microscopy, Electron, Transmission , RNA, Viral , SARS-CoV-2/physiology , Virion/ultrastructure , Virus Assembly , Virus Replication
13.
Pathobiology ; 88(1): 69-77, 2021.
Article in English | MEDLINE | ID: covidwho-781257

ABSTRACT

Since the outbreak of coronavirus disease 2019 (COVID-19), there has been a debate whether pregnant women are at a specific risk for COVID-19 and whether it might be vertically transmittable through the placenta. We present a series of five placentas of SARS coronavirus 2 (SARS-CoV-2)-positive women who had been diagnosed with mild symptoms of COVID-19 or had been asymptomatic before birth. We provide a detailed histopathologic description of morphological changes accompanied by an analysis of presence of SARS-CoV-2 in the placental tissue. All placentas were term deliveries (40th and 41st gestational weeks). One SARS-CoV-2-positive patient presented with cough and dyspnoea. This placenta showed prominent lymphohistiocytic villitis and intervillositis and signs of maternal and foetal malperfusion. Viral RNA was present in both placenta tissue and the umbilical cord and could be visualized by in situ hybridization in the decidua. SARS-CoV-2 tests were negative at the time of delivery of 3/5 women, and their placentas did not show increased inflammatory infiltrates. Signs of maternal and/or foetal malperfusion were present in 100% and 40% of cases, respectively. There was no transplacental transmission to the infants. In our cohort, we can document different time points regarding SARS-CoV-2 infection. In acute COVID-19, prominent lymphohistiocytic villitis may occur and might potentially be attributable to SARS-CoV-2 infection of the placenta. Furthermore, there are histopathological signs of maternal and foetal malperfusion, which might have a relationship to an altered coagulative or microangiopathic state induced by SARS-CoV-2, yet this cannot be proven considering a plethora of confounding factors.


Subject(s)
COVID-19/pathology , COVID-19/virology , Placenta/virology , SARS-CoV-2/pathogenicity , Adult , Cohort Studies , Female , Humans , Placenta/pathology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL