Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Int Immunopharmacol ; 120: 110240, 2023 Jul.
Article in English | MEDLINE | ID: covidwho-2313874

ABSTRACT

Pneumonia is an acute inflammation of the lungs induced by pathogenic microorganisms, immune damage, physical and chemical factors, and other factors, and the latest outbreak of novel coronavirus pneumonia is also an acute lung injury (ALI) induced by viral infection. However, there are currently no effective treatments for inflammatory cytokine storms in patients with ALI/acute respiratory distress syndrome (ARDS). Protein kinase D (PKD) is a highly active kinase that has been shown to be associated with the production of inflammatory cytokines. Therefore, small-molecule compounds that inhibit PKD may be potential drugs for the treatment of ALI/ARDS. In the present study, we evaluated the ability of the small-molecule inhibitor CRT0066101 to attenuate lipopolysaccharide (LPS)-induced inflammatory cytokine production through in vitro cell experiments and a mouse pneumonia model. We found that CRT0066101 significantly reduced the protein and mRNA levels of LPS-induced cytokines (e.g., IL-6, TNF-α, and IL-1ß). CRT0066101 inhibited MyD88 and TLR4 expression and reduced NF-κB, ERK, and JNK phosphorylation. CRT0066101 also reduced NLRP3 activation, inhibited the assembly of the inflammasome complex, and attenuated inflammatory cell infiltration and lung tissue damage. Taken together, our data indicate that CRT0066101 exerts anti-inflammatory effects on LPS-induced inflammation through the TLR4/MyD88 signaling pathway, suggesting that CRT0066101 may have therapeutic value in acute lung injury and other MyD88-dependent inflammatory diseases.


Subject(s)
Acute Lung Injury , COVID-19 , Pneumonia , Respiratory Distress Syndrome , Mice , Animals , Cytokine Release Syndrome/metabolism , Myeloid Differentiation Factor 88/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/metabolism , COVID-19/metabolism , Lung/pathology , Pneumonia/pathology , Acute Lung Injury/chemically induced , NF-kappa B/metabolism , Inflammation/metabolism , Cytokines/metabolism , Respiratory Distress Syndrome/metabolism
2.
PLoS One ; 18(5): e0285770, 2023.
Article in English | MEDLINE | ID: covidwho-2319825

ABSTRACT

Pneumonia, always a major malady, became the main public health and economic disaster of historical proportions with the COVID-19 pandemic. This study was based on a premise that pathology of lung metabolism in inflammation may have features invariant to the nature of the underlying cause. Amino acid uptake by the lungs was measured from plasma samples collected pre-terminally from a carotid artery and vena cava in mice with bleomycin-induced lung inflammation (N = 10) and compared to controls treated with saline instillation (N = 6). In the control group, the difference in concentrations between the arterial and venous blood of the 19 amino acids measured reached the level of statistical significance only for arginine (-10.7%, p = 0.0372) and phenylalanine (+5.5%, p = 0.0266). In the bleomycin group, 11 amino acids had significantly lower concentrations in the arterial blood. Arginine concentration was decreased by 21.1% (p<0.0001) and only that of citrulline was significantly increased (by 20.1%, p = 0.0002). Global Arginine Bioavailability Ratio was decreased in arterial blood by 19.5% (p = 0.0305) in the saline group and by 30.4% (p<0.0001) in the bleomycin group. Production of nitric oxide (NO) and citrulline from arginine by the inducible nitric oxide synthase (iNOS) is greatly increased in the immune system's response to lung injury. Deprived of arginine, the endothelial cells downstream may fail to provide enough NO to prevent the activation of thrombocytes. Thrombotic-related vascular dysfunction is a defining characteristic of pneumonia, including COVID-19. This experiment lends further support to arginine replacement as adjuvant therapy in pneumonia.


Subject(s)
COVID-19 , Pneumonia , Mice , Humans , Animals , Arginine/metabolism , Bleomycin/toxicity , Endothelial Cells/metabolism , Citrulline/metabolism , Pandemics , COVID-19/pathology , Lung/pathology , Pneumonia/pathology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism
3.
Wiley Interdiscip Rev Nanomed Nanobiotechnol ; 14(2): e1763, 2022 03.
Article in English | MEDLINE | ID: covidwho-2173486

ABSTRACT

Pneumonia is a common but serious infectious disease, and is the sixth leading cause for death. The foreign pathogens such as viruses, fungi, and bacteria establish an inflammation response after interaction with lung, leading to the filling of bronchioles and alveoli with fluids. Although the pharmacotherapies have shown their great effectiveness to combat pathogens, advanced methods are under developing to treat complicated cases such as virus-infection and lung inflammation or acute lung injury (ALI). The inflammation modulation nanoparticles (NPs) can effectively suppress immune cells and inhibit inflammatory molecules in the lung site, and thereby alleviate pneumonia and ALI. In this review, the pathological inflammatory microenvironments in pneumonia, which are instructive for the design of biomaterials therapy, are summarized. The focus is then paid to the inflammation-modulating NPs that modulate the inflammatory cells, cytokines and chemokines, and microenvironments of pneumonia for better therapeutic effects. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Respiratory Disease.


Subject(s)
Acute Lung Injury , Nanoparticles , Pneumonia , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Humans , Inflammation/drug therapy , Lung , Nanoparticles/therapeutic use , Pneumonia/drug therapy , Pneumonia/pathology
4.
Int J Mol Sci ; 23(24)2022 Dec 13.
Article in English | MEDLINE | ID: covidwho-2200321

ABSTRACT

Acute respiratory distress syndrome (ARDS) and sepsis are risk factors contributing to mortality in patients with pneumonia. In ARDS, also termed acute lung injury (ALI), pulmonary immune responses lead to excessive pro-inflammatory cytokine release and aberrant alveolar neutrophil infiltration. Systemic spread of cytokines is associated with systemic complications including sepsis, multi-organ failure, and death. Thus, dampening pro-inflammatory cytokine release is a viable strategy to improve outcome. Activation of cannabinoid type II receptor (CB2) has been shown to reduce cytokine release in various in vivo and in vitro studies. Herein, we investigated the effect of HU-308, a specific CB2 agonist, on systemic and pulmonary inflammation in a model of pneumonia-induced ALI. C57Bl/6 mice received intranasal endotoxin or saline, followed by intravenous HU-308, dexamethasone, or vehicle. ALI was scored by histology and plasma levels of select inflammatory mediators were assessed by Luminex assay. Intravital microscopy (IVM) was performed to assess leukocyte adhesion and capillary perfusion in intestinal and pulmonary microcirculation. HU-308 and dexamethasone attenuated LPS-induced cytokine release and intestinal microcirculatory impairment. HU-308 modestly reduced ALI score, while dexamethasone abolished it. These results suggest administration of HU-308 can reduce systemic inflammation without suppressing pulmonary immune response in pneumonia-induced ALI and systemic inflammation.


Subject(s)
Acute Lung Injury , Cannabinoids , Pneumonia , Respiratory Distress Syndrome , Sepsis , Mice , Animals , Endotoxins/adverse effects , Microcirculation , Pneumonia/drug therapy , Pneumonia/etiology , Pneumonia/pathology , Inflammation/pathology , Lung/pathology , Cannabinoids/adverse effects , Acute Lung Injury/etiology , Acute Lung Injury/chemically induced , Cytokines , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/etiology , Lipopolysaccharides/toxicity , Dexamethasone/adverse effects , Mice, Inbred C57BL
5.
Medicine (Baltimore) ; 101(39): e30744, 2022 Sep 30.
Article in English | MEDLINE | ID: covidwho-2113766

ABSTRACT

OBJECTIVE: The aim of this study was to compare the radiographic features of patients with progressive and nonprogressive coronavirus disease 2019 (COVID-19) pneumonia. METHODS: PubMed, Embase, and Cochrane Library databases were searched from January 1, 2020, to February 28, 2022, by using the keywords: "COVID-19", "novel Coronavirus", "2019-novel coronavirus", "CT", "radiology" and "imaging". We summarized the computed tomography manifestations of progressive and nonprogressive COVID-19 pneumonia. The meta-analysis was performed using the Stata statistical software version 16.0. RESULTS: A total of 10 studies with 1092 patients were included in this analysis. The findings of this meta-analysis indicated that the dominating computed tomography characteristics of progressive patients were a crazy-paving pattern (odds ratio [OR] = 2.10) and patchy shadowing (OR = 1.64). The dominating lesions distribution of progressive patients were bilateral (OR = 11.62), central mixed subpleural (OR = 1.37), and central (OR = 1.36). The other dominating lesions of progressive patients were pleura thickening (OR = 2.13), lymphadenopathy (OR = 1.74), vascular enlargement (OR = 1.39), air bronchogram (OR = 1.29), and pleural effusion (OR = 1.29). Two patterns of lesions showed significant links with the progression of disease: nodule (P = .001) and crazy-paving pattern (P = .023). Four lesions distribution showed significant links with the progression of disease: bilateral (P = .004), right upper lobe (P = .003), right middle lobe (P = .001), and left upper lobe (P = .018). CONCLUSION: Nodules, crazy-paving pattern, and/or new lesions in bilateral, upper and middle lobe of right lung, and lower lobe of left lung may indicate disease deterioration. Clinicians should formulate or modify treatment strategies in time according to these specific conditions.


Subject(s)
COVID-19 , Pneumonia , COVID-19/diagnostic imaging , Humans , Lung/diagnostic imaging , Lung/pathology , Pneumonia/pathology , Retrospective Studies , SARS-CoV-2 , Tomography, X-Ray Computed/methods
6.
Biomed Res Int ; 2022: 3510423, 2022.
Article in English | MEDLINE | ID: covidwho-2020494

ABSTRACT

Purpose: Aurantiamide acetate (AA) is a dipeptide derivative with complex pharmacological activities and remarkable effects on preventing and treating various diseases. In the current study, we aimed to investigate whether AA can exert protective effects in a mouse model of ALI induced by LPS. Materials and Methods: In this model, mice were given intranasal LPS for 3 days prior to receiving AA (2.5, 5, and 10 mg/kg) via oral gavage. An assessment of histopathological changes was performed by hematoxylin and eosin (HE). Proinflammatory cytokines were detected in bronchoalveolar lavage fluids (BALFs) by enzyme-linked immunosorbent assays (ELISAs). The effects of AA on protein expression of NF-κB and PI3K/AKT signaling pathways were determined by Western blot. In addition, lung wet/dry (W/D) weight ratio, myeloperoxidase (MPO) activity, cell counts, and protein content were also measured. Results: According to results, AA pretreatment significantly reduced lung pathological changes, W/D ratio, MPO activity, and protein content. Additionally, AA resulted in a significant reduction in the number of total cells, neutrophils, and proinflammatory cytokines in the BALF after LPS stimulation. The subsequent study revealed that pretreatment with AA dose dependently suppressed LPS-induced activation of NF-κB as well as PI3K/AKT phosphorylation. Conclusion: The results indicated that the AA had a protective effect on LPS-induced ALI in mice and could be a potential drug for ALI.


Subject(s)
Acute Lung Injury , Pneumonia , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/metabolism , Dipeptides/pharmacology , Lipopolysaccharides/adverse effects , Lung/pathology , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pneumonia/pathology , Proto-Oncogene Proteins c-akt/metabolism
7.
PLoS One ; 17(7): e0272019, 2022.
Article in English | MEDLINE | ID: covidwho-1963043

ABSTRACT

Coronavirus disease (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is currently spreading globally. To overcome the COVID-19 pandemic, preclinical evaluations of vaccines and therapeutics using K18-hACE2 and CAG-hACE2 transgenic mice are ongoing. However, a comparative study on SARS-CoV-2 infection between K18-hACE2 and CAG-hACE2 mice has not been published. In this study, we compared the susceptibility and resistance to SARS-CoV-2 infection between two strains of transgenic mice, which were generated in FVB background mice. K18-hACE2 mice exhibited severe weight loss with definitive lethality, but CAG-hACE2 mice survived; and differences were observed in the lung, spleen, cerebrum, cerebellum, and small intestine. A higher viral titer was detected in the lungs, cerebrums, and cerebellums of K18-hACE2 mice than in the lungs of CAG-hACE2 mice. Severe pneumonia was observed in histopathological findings in K18-hACE2, and mild pneumonia was observed in CAG-hACE2. Atrophy of the splenic white pulp and reduction of spleen weight was observed, and hyperplasia of goblet cells with villi atrophy of the small intestine was observed in K18-hACE2 mice compared to CAG-hACE2 mice. These results indicate that K18-hACE2 mice are relatively susceptible to SARS-CoV-2 and that CAG-hACE2 mice are resistant to SARS-CoV-2. Based on these lineage-specific sensitivities, we suggest that K18-hACE2 mouse is suitable for highly susceptible model of SARS-CoV-2, and CAG-hACE2 mouse is suitable for mild susceptible model of SARS-CoV-2 infection.


Subject(s)
COVID-19 , Pneumonia , Angiotensin-Converting Enzyme 2/genetics , Animals , Atrophy/pathology , Disease Models, Animal , Disease Susceptibility/pathology , Humans , Lung/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Pandemics , Peptidyl-Dipeptidase A , Pneumonia/pathology , SARS-CoV-2
8.
Crit Care ; 26(1): 155, 2022 05 30.
Article in English | MEDLINE | ID: covidwho-1951297

ABSTRACT

BACKGROUND: A dysregulated immune response is emerging as a key feature of critical illness in COVID-19. Neutrophils are key components of early innate immunity that, if not tightly regulated, contribute to uncontrolled systemic inflammation. We sought to decipher the role of neutrophil phenotypes, functions, and homeostasis in COVID-19 disease severity and outcome. METHODS: By using flow cytometry, this longitudinal study compares peripheral whole-blood neutrophils from 90 COVID-19 ICU patients with those of 22 SARS-CoV-2-negative patients hospitalized for severe community-acquired pneumonia (CAP) and 38 healthy controls. We also assessed correlations between these phenotypic and functional indicators and markers of endothelial damage as well as disease severity. RESULTS: At ICU admission, the circulating neutrophils of the COVID-19 patients showed continuous basal hyperactivation not seen in CAP patients, associated with higher circulating levels of soluble E- and P-selectin, which reflect platelet and endothelial activation. Furthermore, COVID-19 patients had expanded aged-angiogenic and reverse transmigrated neutrophil subsets-both involved in endothelial dysfunction and vascular inflammation. Simultaneously, COVID-19 patients had significantly lower levels of neutrophil oxidative burst in response to bacterial formyl peptide. Moreover patients dying of COVID-19 had significantly higher expansion of aged-angiogenic neutrophil subset and greater impairment of oxidative burst response than survivors. CONCLUSIONS: These data suggest that neutrophil exhaustion may be involved in the pathogenesis of severe COVID-19 and identify angiogenic neutrophils as a potentially harmful subset involved in fatal outcome.


Subject(s)
COVID-19 , Community-Acquired Infections , Pneumonia , Aged , Homeostasis , Humans , Inflammation , Longitudinal Studies , Neutrophils/physiology , Pneumonia/pathology , SARS-CoV-2 , Severity of Illness Index
9.
Int J Mol Sci ; 21(9)2020 Apr 30.
Article in English | MEDLINE | ID: covidwho-1934078

ABSTRACT

Severe acute respiratory syndrome-associated coronavirus (SARS-CoV) initiates the cytokine/chemokine storm-mediated lung injury. The SARS-CoV unique domain (SUD) with three macrodomains (N, M, and C), showing the G-quadruplex binding activity, was examined the possible role in SARS pathogenesis in this study. The chemokine profile analysis indicated that SARS-CoV SUD significantly up-regulated the expression of CXCL10, CCL5 and interleukin (IL)-1ß in human lung epithelial cells and in the lung tissues of the mice intratracheally instilled with the recombinant plasmids. Among the SUD subdomains, SUD-MC substantially activated AP-1-mediated CXCL10 expression in vitro. In the wild type mice, SARS-CoV SUD-MC triggered the pulmonary infiltration of macrophages and monocytes, inducing CXCL10-mediated inflammatory responses and severe diffuse alveolar damage symptoms. Moreover, SUD-MC actuated NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome-dependent pulmonary inflammation, as confirmed by the NLRP3 inflammasome inhibitor and the NLRP3-/- mouse model. This study demonstrated that SARS-CoV SUD modulated NLRP3 inflammasome-dependent CXCL10-mediated pulmonary inflammation, providing the potential therapeutic targets for developing the antiviral agents.


Subject(s)
Chemokine CXCL10/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Proteins/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Chemokine CXCL10/genetics , Disease Models, Animal , Humans , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pneumonia/pathology , Pneumonia/virology , Promoter Regions, Genetic , Severe acute respiratory syndrome-related coronavirus/isolation & purification , Severe Acute Respiratory Syndrome/pathology , Severe Acute Respiratory Syndrome/virology , Up-Regulation , Viral Proteins/chemistry , Viral Proteins/genetics
10.
J Biomed Sci ; 29(1): 52, 2022 Jul 11.
Article in English | MEDLINE | ID: covidwho-1928188

ABSTRACT

BACKGROUND: Coronavirus-induced disease 19 (COVID-19) infects more than three hundred and sixty million patients worldwide, and people with severe symptoms frequently die of acute respiratory distress syndrome (ARDS). Recent studies indicated that excessive neutrophil extracellular traps (NETs) contributed to immunothrombosis, thereby leading to extensive intravascular coagulopathy and multiple organ dysfunction. Thus, understanding the mechanism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced NET formation would be helpful to reduce thrombosis and prevent ARDS in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. METHODS: We incubated SARS-CoV-2 with neutrophils in the presence or absence of platelets to observe NET formation. We further isolated extracellular vesicles from COVID-19 patients' sera (COVID-19-EVs) to examine their ability to induce NET formation. RESULTS: We demonstrated that antagonistic mAbs against anti-CLEC5A mAb and anti-TLR2 mAb can inhibit COVID-19-EVs-induced NET formation, and generated clec5a-/-/tlr2-/- mice to confirm the critical roles of CLEC5A and TLR2 in SARS-CoV-2-induced lung inflammation in vivo. We found that virus-free extracellular COVID-19 EVs induced robust NET formation via Syk-coupled C-type lectin member 5A (CLEC5A) and TLR2. Blockade of CLEC5A inhibited COVID-19 EVs-induced NETosis, and simultaneous blockade of CLEC5A and TLR2 further suppressed SARS-CoV-2-induced NETosis in vitro. Moreover, thromboinflammation was attenuated dramatically in clec5a-/-/tlr2-/- mice. CONCLUSIONS: This study demonstrates that SARS-CoV-2-activated platelets produce EVs to enhance thromboinflammation via CLEC5A and TLR2, and highlight the importance of CLEC5A and TLR2 as therapeutic targets to reduce the risk of ARDS in COVID-19 patients.


Subject(s)
COVID-19 , Lectins, C-Type , Neutrophils , Pneumonia , Respiratory Distress Syndrome , SARS-CoV-2 , Thrombosis , Animals , Blood Platelets/immunology , Blood Platelets/pathology , Blood Platelets/virology , COVID-19/blood , COVID-19/immunology , Humans , Lectins, C-Type/immunology , Mice , Neutrophils/immunology , Neutrophils/pathology , Neutrophils/virology , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/virology , Receptors, Cell Surface , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/virology , SARS-CoV-2/immunology , Thrombosis/blood , Thrombosis/immunology , Thrombosis/virology , Toll-Like Receptor 2/immunology
11.
Nucl Med Biol ; 112-113: 1-8, 2022.
Article in English | MEDLINE | ID: covidwho-1867632

ABSTRACT

RATIONALE: The aim of this study was to investigate the application of [18F]DPA714 to visualize the inflammation process in the lungs of SARS-CoV-2-infected rhesus monkeys, focusing on the presence of pulmonary lesions, activation of mediastinal lymph nodes and surrounded lung tissue. METHODS: Four experimentally SARS-CoV-2 infected rhesus monkeys were followed for seven weeks post infection (pi) with a weekly PET-CT using [18F]DPA714. Two PET images, 10 min each, of a single field-of-view covering the chest area, were obtained 10 and 30 min after injection. To determine the infection process swabs, blood and bronchoalveolar lavages (BALs) were obtained. RESULTS: All animals were positive for SARS-CoV-2 in both the swabs and BALs on multiple timepoints pi. The initial development of pulmonary lesions was already detected at the first scan, performed 2-days pi. PET revealed an increased tracer uptake in the pulmonary lesions and mediastinal lymph nodes of all animals from the first scan obtained after infection and onwards. However, also an increased uptake was detected in the lung tissue surrounding the lesions, which persisted until day 30 and then subsided by day 37-44 pi. In parallel, a similar pattern of increased expression of activation markers was observed on dendritic cells in blood. PRINCIPAL CONCLUSIONS: This study illustrates that [18F]DPA714 is a valuable radiotracer to visualize SARS-CoV-2-associated pulmonary inflammation, which coincided with activation of dendritic cells in blood. [18F]DPA714 thus has the potential to be of added value as diagnostic tracer for other viral respiratory infections.


Subject(s)
COVID-19 , Pneumonia , Animals , COVID-19/diagnostic imaging , Lung/diagnostic imaging , Lung/pathology , Macaca mulatta , Pneumonia/diagnostic imaging , Pneumonia/pathology , Positron Emission Tomography Computed Tomography/methods , Pyrazoles , Pyrimidines , SARS-CoV-2
12.
Chem Biol Interact ; 362: 109982, 2022 Aug 01.
Article in English | MEDLINE | ID: covidwho-1850754

ABSTRACT

In this Letter to the Editor supportive data were presented to a recent paper published in this journal reporting the involvement of TRP channels in COVID-19 pneumonia and its role for new therapies. Since gene expression of TRP channels was found in human lung tissues the protein was not being reported so far. TRP channels are supposed to be involved in the pulmonary inflammation and its symptoms such as fever, cough and others. Here, TRPC6 was investigated in tissues of normal human lungs and of SARS-Cov-2 infected lungs in a preliminary study. Tissue was obtained post mortem from anatomical body donations during dissections and during pathological dissections (13 normal, 4 COVID-19 pneumoniae) and processed for immunohistochemistry. In normal lungs TRPC6 was found in the ciliated epithelium, in the wall of larger lung vessels and in the alveolar septa. In COVID-19 pneumonia the distribution of TRPC6 was different. Inflammatory lesions, cellular infiltrates, hyaline membranes and fibrosis were labelled intensively as well as dilated capillaries. These observations are from four patients with COVID-19 pneumonia.The observations do not elucidate the molecular mechanisms but support the view that TRPC6 channels are involved in normal physiology of normal human lungs and in COVID-19 pneumonia. TRPC6 might aggravate SARS-2 induced inflammation and could be a target for inhibiting drugs.


Subject(s)
COVID-19 , Pneumonia , Humans , Lung/pathology , Pneumonia/metabolism , Pneumonia/pathology , SARS-CoV-2 , TRPC6 Cation Channel/metabolism
13.
Malays J Pathol ; 44(1): 83-92, 2022 Apr.
Article in English | MEDLINE | ID: covidwho-1812960

ABSTRACT

INTRODUCTION: Data on pathological changes in COVID-19 are scarce. The aim of this study was to describe the histopathological and virological findings of postmortem biopsies, and the existing clinical correlations, in people who died of COVID-19. MATERIALS AND METHODS: We performed postmortem needle core biopsies of the chest in 11 people who died of COVID-19 pneumonia. Tissue examination was done by light microscopy and real-time polymerase chain reaction (RTPCR). RESULTS: The age of the patients were between 61 to 94 years. Of the 11 postmortem chest biopsies, lung tissue was obtained in 8, myocardium tissue in 7, and liver tissue in 5. Histologically of lung, the main findings pertaining to the lung were diffuse alveolar damage in proliferative phase (n = 4, 50%), diffuse alveolar damage in exudative and proliferative phase (n = 3, 37.5%), diffuse alveolar damage in exudative (n=1; 12.5%) and acute pneumonia (n = 2, 25%). Necrotising pneumonia, acute fibrinous and organising pneumonia, and neutrophils were detected in one sample each (12.5%). Another case presented myocarditis. RT-PCR showed RNA of SARS-CoV-2 in 7 of the 8 lung samples (87.5%), 2 of the 7 myocardial tissue samples (28.6%), and 1 of the 5 liver tissue samples (20%). CONCLUSION: The postmortem examinations show diffuse alveolar damage, as well as acute or necrotising pneumonia. RT-PCR of SARS-CoV-2 was positive in most lung samples.


Subject(s)
COVID-19 , Pneumonia, Necrotizing , Pneumonia , Aged , Aged, 80 and over , Biopsy, Needle , Humans , Liver/pathology , Lung/pathology , Middle Aged , Pneumonia/pathology , Pneumonia, Necrotizing/pathology , SARS-CoV-2
14.
Sci Rep ; 12(1): 4270, 2022 03 11.
Article in English | MEDLINE | ID: covidwho-1740475

ABSTRACT

Inflammatory burden is associated with COVID-19 severity and outcomes. Residual computed tomography (CT) lung abnormalities have been reported after COVID-19. The aim was to evaluate the association between inflammatory burden during COVID-19 and residual lung CT abnormalities collected on follow-up CT scans performed 2-3 and 6-7 months after COVID-19, in severe COVID-19 pneumonia survivors. C-reactive protein (CRP) curves describing inflammatory burden during the clinical course were built, and CRP peaks, velocities of increase, and integrals were calculated. Other putative determinants were age, sex, mechanical ventilation, lowest PaO2/FiO2 ratio, D-dimer peak, and length of hospital stay (LOS). Of the 259 included patients (median age 65 years; 30.5% females), 202 (78%) and 100 (38.6%) had residual, predominantly non-fibrotic, abnormalities at 2-3 and 6-7 months, respectively. In age- and sex-adjusted models, best CRP predictors for residual abnormalities were CRP peak (odds ratio [OR] for one standard deviation [SD] increase = 1.79; 95% confidence interval [CI] = 1.23-2.62) at 2-3 months and CRP integral (OR for one SD increase = 2.24; 95%CI = 1.53-3.28) at 6-7 months. Hence, inflammation is associated with short- and medium-term lung damage in COVID-19. Other severity measures, including mechanical ventilation and LOS, but not D-dimer, were mediators of the relationship between CRP and residual abnormalities.


Subject(s)
COVID-19/pathology , Pneumonia/diagnostic imaging , Aged , C-Reactive Protein/analysis , COVID-19/complications , COVID-19/diagnostic imaging , Female , Humans , Male , Middle Aged , Patient Acuity , Pneumonia/etiology , Pneumonia/pathology , Retrospective Studies , Risk Factors , Time Factors , Tomography, X-Ray Computed
15.
J Immunol ; 208(2): 321-327, 2022 01 15.
Article in English | MEDLINE | ID: covidwho-1708204

ABSTRACT

Previous studies have demonstrated that 8-hydroxydeoxyguanosine (8-OHdG) exerted key roles in various pulmonary diseases, but the evidence for its role in community-acquired pneumonia (CAP) was lacking. The goal of this research was to evaluate the correlations of serum 8-OHdG with the severity and prognosis among patients with CAP through a prospective cohort study. A total of 239 patients with CAP and 239 healthy participants were enrolled. Fasting blood samples were collected. 8-OHdG and inflammatory cytokines were measured by ELISA. On admission, serum 8-OHdG was significantly increased in patients with CAP compared with control subjects. Besides, serum 8-OHdG was incrementally increased in line with CAP severity scores. Pearson correlative analysis found that serum 8-OHdG was correlated with clinical characteristics and inflammatory cytokines in patients with CAP. Linear and logistic regression analysis showed that serum 8-OHdG was positively associated with CAP severity scores. Furthermore, the prognostic outcomes were tracked. Higher serum 8-OHdG on admission increased the risks for intensive care unit admission, mechanical ventilation, vasoactive agent usage, death, and longer hospital stay among patients with CAP. Serum 8-OHdG combination with confusion, respiratory rate, blood pressure, and age ≥65 y or pneumonia severity index had stronger predictive powers for death than single 8-OHdG, CAP severity scores, or several inflammatory cytokines in patients with CAP. These results indicated that serum 8-OHdG is positively associated with the severity and poor prognosis in patients with CAP, demonstrating that 8-OHdG may be involved in the pathophysiology process of CAP.


Subject(s)
8-Hydroxy-2'-Deoxyguanosine/blood , Community-Acquired Infections/pathology , Pneumonia/blood , Pneumonia/mortality , Severity of Illness Index , Aged , Biomarkers/blood , Community-Acquired Infections/blood , Critical Care/statistics & numerical data , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Length of Stay/statistics & numerical data , Male , Middle Aged , Oxidative Stress/physiology , Pneumonia/pathology , Prognosis , Prospective Studies , Respiration, Artificial/statistics & numerical data
16.
Nature ; 603(7899): 145-151, 2022 03.
Article in English | MEDLINE | ID: covidwho-1631700

ABSTRACT

COVID-19, which is caused by infection with SARS-CoV-2, is characterized by lung pathology and extrapulmonary complications1,2. Type I interferons (IFNs) have an essential role in the pathogenesis of COVID-19 (refs 3-5). Although rapid induction of type I IFNs limits virus propagation, a sustained increase in the levels of type I IFNs in the late phase of the infection is associated with aberrant inflammation and poor clinical outcome5-17. Here we show that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, which controls immunity to cytosolic DNA, is a critical driver of aberrant type I IFN responses in COVID-19 (ref. 18). Profiling COVID-19 skin manifestations, we uncover a STING-dependent type I IFN signature that is primarily mediated by macrophages adjacent to areas of endothelial cell damage. Moreover, cGAS-STING activity was detected in lung samples from patients with COVID-19 with prominent tissue destruction, and was associated with type I IFN responses. A lung-on-chip model revealed that, in addition to macrophages, infection with SARS-CoV-2 activates cGAS-STING signalling in endothelial cells through mitochondrial DNA release, which leads to cell death and type I IFN production. In mice, pharmacological inhibition of STING reduces severe lung inflammation induced by SARS-CoV-2 and improves disease outcome. Collectively, our study establishes a mechanistic basis of pathological type I IFN responses in COVID-19 and reveals a principle for the development of host-directed therapeutics.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Interferon Type I/immunology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , SARS-CoV-2/immunology , Animals , COVID-19/metabolism , COVID-19/virology , Cells, Cultured , DNA, Mitochondrial/metabolism , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Female , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Macrophages/immunology , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/virology , SARS-CoV-2/pathogenicity , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/pathology
17.
Int J Mol Sci ; 23(3)2022 Jan 19.
Article in English | MEDLINE | ID: covidwho-1625612

ABSTRACT

Repurposing of the anthelminthic drug niclosamide was proposed as an effective treatment for inflammatory airway diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease. Niclosamide may also be effective for the treatment of viral respiratory infections, such as SARS-CoV-2, respiratory syncytial virus, and influenza. While systemic application of niclosamide may lead to unwanted side effects, local administration via aerosol may circumvent these problems, particularly when the drug is encapsulated into small polyethylene glycol (PEG) hydrospheres. In the present study, we examined whether PEG-encapsulated niclosamide inhibits the production of mucus and affects the pro-inflammatory mediator CLCA1 in mouse airways in vivo, while effects on mucociliary clearance were assessed in excised mouse tracheas. The potential of encapsulated niclosamide to inhibit TMEM16A whole-cell Cl- currents and intracellular Ca2+ signalling was assessed in airway epithelial cells in vitro. We achieved encapsulation of niclosamide in PEG-microspheres and PEG-nanospheres (Niclo-spheres). When applied to asthmatic mice via intratracheal instillation, Niclo-spheres strongly attenuated overproduction of mucus, inhibited secretion of the major proinflammatory mediator CLCA1, and improved mucociliary clearance in tracheas ex vivo. These effects were comparable for niclosamide encapsulated in PEG-nanospheres and PEG-microspheres. Niclo-spheres inhibited the Ca2+ activated Cl- channel TMEM16A and attenuated mucus production in CFBE and Calu-3 human airway epithelial cells. Both inhibitory effects were explained by a pronounced inhibition of intracellular Ca2+ signals. The data indicate that poorly dissolvable compounds such as niclosamide can be encapsulated in PEG-microspheres/nanospheres and deposited locally on the airway epithelium as encapsulated drugs, which may be advantageous over systemic application.


Subject(s)
Niclosamide/administration & dosage , Pneumonia/drug therapy , Respiratory System/drug effects , Animals , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , COVID-19/complications , Cells, Cultured , Disease Models, Animal , Drug Carriers/chemistry , Drug Compounding , Humans , Hydrogels/chemistry , Instillation, Drug , Mice , Microspheres , Mucus/drug effects , Mucus/metabolism , Nanospheres/administration & dosage , Nanospheres/chemistry , Niclosamide/chemistry , Niclosamide/pharmacokinetics , Pneumonia/pathology , Polyethylene Glycols/chemistry , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory System/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Trachea , COVID-19 Drug Treatment
18.
Front Immunol ; 12: 784028, 2021.
Article in English | MEDLINE | ID: covidwho-1581324

ABSTRACT

Background: Extracellular vesicles (EVs) are mediators of cell-to-cell communication in inflammatory lung diseases. They function as carriers for miRNAs which regulate mRNA transcripts and signaling pathways after uptake into recipient cells. We investigated whether miRNAs associated with circulating EVs regulate immunologic processes in COVID-19. Methods: We prospectively studied 20 symptomatic patients with COVID-19 pneumonia, 20 mechanically ventilated patients with severe COVID-19 (severe acute respiratory corona virus-2 syndrome, ARDS) and 20 healthy controls. EVs were isolated by precipitation, total RNA was extracted, profiled by small RNA sequencing and evaluated by differential gene expression analysis (DGE). Differentially regulated miRNAs between groups were bioinformatically analyzed, mRNA target transcripts identified and signaling networks constructed, thereby comparing COVID-19 pneumonia to the healthy state and pneumonia to severe COVID-19 ARDS. Results: DGE revealed 43 significantly and differentially expressed miRNAs (25 downregulated) in COVID-19 pneumonia when compared to controls, and 20 miRNAs (15 downregulated) in COVID-19 ARDS patients in comparison to those with COVID-19 pneumonia. Network analysis for comparison of COVID-19 pneumonia to healthy controls showed upregulated miR-3168 (log2FC=2.28, padjusted<0.001), among others, targeting interleukin-6 (IL6) (25.1, 15.2 - 88.2 pg/ml in COVID-19 pneumonia) and OR52N2, an olfactory smell receptor in the nasal epithelium. In contrast, miR-3168 was significantly downregulated in COVID-19 ARDS (log2FC=-2.13, padjusted=0.003) and targeted interleukin-8 (CXCL8) in a completely activated network. Toll-like receptor 4 (TLR4) was inhibited in COVID-19 pneumonia by miR-146a-5p and upregulated in ARDS by let-7e-5p. Conclusion: EV-derived miRNAs might have important regulative functions in the pathophysiology of COVID-19: CXCL8 regulates neutrophil recruitment into the lung causing epithelial damage whereas activated TLR4, to which SARS-CoV-2 spike protein binds strongly, increases cell surface ACE2 expression and destroys type II alveolar cells that secrete pulmonary surfactants; both resulting in pulmonary-capillary leakage and ARDS. These miRNAs may serve as biomarkers or as possible therapeutic targets.


Subject(s)
Biomarkers/blood , COVID-19/immunology , Extracellular Vesicles/immunology , MicroRNAs/immunology , Aged , Aged, 80 and over , COVID-19/pathology , Disease Progression , Female , Humans , Male , Middle Aged , Pneumonia/immunology , Pneumonia/pathology , SARS-CoV-2 , Signal Transduction/immunology
19.
J Med Virol ; 94(4): 1289-1291, 2022 Apr.
Article in English | MEDLINE | ID: covidwho-1589037

ABSTRACT

In December 2019, a new type of virus, coronavirus disease 2019 broke out globally and caused great harm. The virus mutates rapidly, and more research reports are urgently needed to increase our understanding of the disease. We found the reversed halo sign (RHS) occurred in the imaging manifestations of severe acute respiratory syndrome coronavirus 2 delta variant of concern pneumonia. In the absence of pathology, the mechanism is unknown. Therefore, we reported two cases of RHS and tried to speculate the pathological mechanism through multiple computed tomography follow-up comparisons to judge the prognosis of the disease.


Subject(s)
COVID-19/diagnostic imaging , SARS-CoV-2/pathogenicity , COVID-19/pathology , COVID-19/virology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pneumonia/diagnostic imaging , Pneumonia/pathology , Pneumonia/virology , Prognosis , SARS-CoV-2/isolation & purification , Tomography, X-Ray Computed
20.
J Ethnopharmacol ; 287: 114965, 2022 Apr 06.
Article in English | MEDLINE | ID: covidwho-1587284

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Coronavirus and influenza virus infection seriously threaten human health. Cangma Huadu Granules (CMHD) is an in-hospital preparation composed of eight traditional Chinese medicines (TCM), which has been clinically used against COVID-19 in China and may be a promising candidate for the treatment of influenza. However, the role of its treatment urgently needs to be studied. AIM OF THE STUDY: To evaluate the therapeutic effects of CMHD on pneumonia induced by coronavirus (HCoV-229E) and influenza A virus (H1N1/FM1) in mice and explore its mechanism of anti-infection. MATERIALS AND METHODS: Mice were infected with HCoV-229E or H1N1/FM1 virus through the nasal cavity. CMHD (12.1, 6.05 and 3.03 g/kg/d) or the positive control drugs were administered intragastrically. The lung index and histopathological changes were used to evaluate the therapeutic effect of CMHD. The expression of TNF-α, IL-1ß, IL-6 and IL-4 in Serum and the proportion of CD4+ and CD8+ T lymphocytes in peripheral blood were detected to evaluate the anti-inflammatory and immune regulation effects of CMHD, respectively. Furthermore, the levels of p-NF-κBp65/ NF-κB p65, which was the key targets of the NF-κB pathway was analyzed. RESULTS: In HCoV-229E-induced pneumonia, the lung index was markedly reduced, and lung pathology was improved in mice that treated with CMHD (12.1, 6.05 g/kg/d). Meanwhile, the expression of TNF-α, IL-6 were obviously inhibited, but the expression of IL-4 was significantly increased in CMHD groups. Compared with the model group, CMHD could also markedly upregulate the level of CD4+ and CD8+. Furthermore, CMHD has a markedly effect on inhibit the expression of p-NF-κB p65/NF-κB p65 in the lung. In H1N1-induced pneumonia, the lung index of mice in the CMHD (12.1 g/kg/d) treatment group was lower than that in the model group, and less inflammatory infiltration could be seen in the lung pathological. Moreover, CMHD could also obviously decrease the expression of TNF-α, IL-1ß, IL-6, but significantly increase the expression of IL-4. Except for that, CMHD could also markedly downregulate the level of CD4+ and upregulate the level of CD8+ compared with the model group. In addition, CMHD has a markedly effect on inhibit the expression of p-NF-κB p65/NF-κB p65 in the lung. CONCLUSION: CMHD can significantly combats viral infections caused by HCoV-229E and H1N1, and the mechanism may be related to its multiple functions of anti-inflammatory, immunity regulating and inhibiting NF-κB signal transduction pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Coronavirus Infections/drug therapy , Drugs, Chinese Herbal/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Medicine, Chinese Traditional/methods , Orthomyxoviridae Infections/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Coronavirus 229E, Human/drug effects , Cytokines/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Female , Immunity/drug effects , Male , Mice, Inbred BALB C , Mice, Inbred ICR , Pneumonia/drug therapy , Pneumonia/pathology , T-Lymphocytes/metabolism , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL