Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 39
Filtre
1.
Aging (Albany NY) ; 15(11): 4625-4641, 2023 06 07.
Article Dans Anglais | MEDLINE | ID: covidwho-20239369

Résumé

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19) pandemic. The severity of COVID-19 increases with each decade of life, a phenomenon that suggest that organismal aging contributes to the fatality of the disease. In this regard, we and others have previously shown that COVID-19 severity correlates with shorter telomeres, a molecular determinant of aging, in patient's leukocytes. Lung injury is a predominant feature of acute SARS-CoV-2 infection that can further progress to lung fibrosis in post-COVID-19 patients. Short or dysfunctional telomeres in Alveolar type II (ATII) cells are sufficient to induce pulmonary fibrosis in mouse and humans. Here, we analyze telomere length and the histopathology of lung biopsies from a cohort of alive post-COVID-19 patients and a cohort of age-matched controls with lung cancer. We found loss of ATII cellularity and shorter telomeres in ATII cells concomitant with a marked increase in fibrotic lung parenchyma remodeling in post- COVID-19 patients compared to controls. These findings reveal a link between presence of short telomeres in ATII cells and long-term lung fibrosis sequel in Post-COVID-19 patients.


Sujets)
COVID-19 , Tumeurs , Fibrose pulmonaire , Humains , Souris , Animaux , Fibrose pulmonaire/anatomopathologie , COVID-19/anatomopathologie , SARS-CoV-2 , Pneumocytes , Poumon/anatomopathologie , Tumeurs/anatomopathologie , Télomère/anatomopathologie
2.
Bioorg Chem ; 138: 106592, 2023 Sep.
Article Dans Anglais | MEDLINE | ID: covidwho-2311750

Résumé

Pulmonary fibrosis is the end-stage change of a large class of lung diseases characterized by the proliferation of fibroblasts and the accumulation of a large amount of extracellular matrix, accompanied by inflammatory damage and tissue structure destruction, which also shows the normal alveolar tissue is damaged and then abnormally repaired resulting in structural abnormalities (scarring). Pulmonary fibrosis has a serious impact on the respiratory function of the human body, and the clinical manifestation is progressive dyspnea. The incidence of pulmonary fibrosis-related diseases is increasing year by year, and no curative drugs have appeared so far. Nevertheless, research on pulmonary fibrosis have also increased in recent years, but there are no breakthrough results. Pathological changes of pulmonary fibrosis appear in the lungs of patients with coronavirus disease 2019 (COVID-19) that have not yet ended, and whether to improve the condition of patients with COVID-19 by means of the anti-fibrosis therapy, which are the questions we need to address now. This review systematically sheds light on the current state of research on fibrosis from multiple perspectives, hoping to provide some references for design and optimization of subsequent drugs and the selection of anti-fibrosis treatment plans and strategies.


Sujets)
COVID-19 , Fibrose pulmonaire , Humains , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , COVID-19/anatomopathologie , Poumon , Fibrose , Fibroblastes
3.
J Appl Microbiol ; 134(1)2023 Jan 23.
Article Dans Anglais | MEDLINE | ID: covidwho-2308562

Résumé

AIMS: To evaluate the effects of the Qingwen Gupi decoction (QGT) in a rat model of bleomycin-induced pulmonary fibrosis (PF), and explore the underlying mechanisms by integrating UPLC-Q-TOF/MS metabolomics and 16S rDNA sequencing of gut microbiota. METHODS AND RESULTS: The animals were randomly divided into the control, PF model, pirfenidone-treated, and low-, medium-, and high-dose QGT groups. The lung tissues were examined and the expression of TGF-ß, SMAD-3, and SMAD-7 mRNAs in the lung tissues were analyzed. Metabolomic profiles were analyzed by UPLC-QTOF/MS, and the intestinal flora were examined by prokaryotic 16 rDNA sequencing. Pathological examination and biochemical indices revealed that QGT treatment improved the symptoms of PF by varying degrees. Furthermore, QGT significantly downregulated TGF-ß1 and Smad-3 mRNAs and increased the expression levels of Smad-7. QGT-L in particular increased the levels of 18 key metabolic biomarkers that were associated with nine gut microbial species and may exert antifibrosis effects through arachidonic acid metabolism, glycerophospholipid metabolism, and phenylalanine metabolism. CONCLUSIONS: QGT alleviated PF in a rat model through its anti-inflammatory, antioxidant, and anti-fibrotic effects, and by reversing bleomycin-induced gut dysbiosis.This study lays the foundation for further research on the pathological mechanisms of PF and the development of new drug candidates.


Sujets)
Microbiome gastro-intestinal , Fibrose pulmonaire , Rats , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Poumon , Bléomycine/effets indésirables , Facteur de croissance transformant bêta/métabolisme , Métabolomique
5.
Front Immunol ; 13: 934264, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-2198854

Résumé

Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), responsible for COVID-19, has caused a global pandemic. Observational studies revealed a condition, herein called as Long-COVID syndrome (PC), that affects both moderately and severely infected patients, reducing quality-of-life. The mechanism/s underlying the onset of fibrotic-like changes in PC are still not well defined. The goal of this study was to understand the involvement of the Absent in melanoma-2 (AIM2) inflammasome in PC-associated lung fibrosis-like changes revealed by chest CT scans. Peripheral blood mononuclear cells (PBMCs) obtained from PC patients who did not develop signs of lung fibrosis were not responsive to AIM2 activation by Poly dA:dT. In sharp contrast, PBMCs from PC patients with signs of lung fibrosis were highly responsive to AIM2 activation, which induced the release of IL-1α, IFN-α and TGF-ß. The recognition of Poly dA:dT was not due to the activation of cyclic GMP-AMP (cGAMP) synthase, a stimulator of interferon response (cGAS-STING) pathways, implying a role for AIM2 in PC conditions. The release of IFN-α was caspase-1- and caspase-4-dependent when AIM2 was triggered. Instead, the release of pro-inflammatory IL-1α and pro-fibrogenic TGF-ß were inflammasome independent because the inhibition of caspase-1 and caspase-4 did not alter the levels of the two cytokines. Moreover, the responsiveness of AIM2 correlated with higher expression of the receptor in circulating CD14+ cells in PBMCs from patients with signs of lung fibrosis.


Sujets)
COVID-19 , Protéines de liaison à l'ADN , Fibrose pulmonaire , COVID-19/sang , COVID-19/immunologie , COVID-19/anatomopathologie , Protéines de transport , Caspase-1/immunologie , Protéines de liaison à l'ADN/sang , Protéines de liaison à l'ADN/immunologie , Humains , Inflammasomes/sang , Inflammasomes/immunologie , Interféron alpha/métabolisme , Agranulocytes/immunologie , Fibrose pulmonaire/sang , Fibrose pulmonaire/immunologie , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/virologie , SARS-CoV-2 , Facteur de croissance transformant bêta/métabolisme ,
7.
J Microbiol Biotechnol ; 30(3): 427-438, 2020 Mar 28.
Article Dans Anglais | MEDLINE | ID: covidwho-2163802

Résumé

Middle East respiratory syndrome coronavirus (MERS-CoV) infects the lower respiratory airway of humans, leading to severe acute respiratory failure. Unlike human dipeptidyl peptidase 4 (hDPP4), a receptor for MERS-CoV, mouse DPP4 (mDPP4) failed to support MERS-CoV infection. Consequently, diverse transgenic mouse models expressing hDPP4 have been developed using diverse methods, although some models show no mortality and/or only transient and mild-to-moderate clinical signs following MERS-CoV infection. Additionally, overexpressed hDPP4 is associated with neurological complications and breeding difficulties in some transgenic mice, resulting in impeding further studies. Here, we generated stable hDPP4-transgenic mice that were sufficiently susceptible to MERS-CoV infection. The transgenic mice showed weight loss, decreased pulmonary function, and increased mortality with minimal perturbation of overexpressed hDPP4 after MERS-CoV infection. In addition, we observed histopathological signs indicative of progressive pulmonary fibrosis, including thickened alveolar septa, infiltration of inflammatory monocytes, and macrophage polarization as well as elevated expression of profibrotic molecules and acute inflammatory response in the lung of MERS-CoV-infected hDPP4-transgenic mice. Collectively, we suggest that this hDPP4-transgenic mouse is useful in understanding the pathogenesis of MERS-CoV infection and for antiviral research and vaccine development against the virus.


Sujets)
Infections à coronavirus/immunologie , Dipeptidyl peptidase 4/immunologie , Poumon/anatomopathologie , Coronavirus du syndrome respiratoire du Moyen-Orient/immunologie , Fibrose pulmonaire/anatomopathologie , Animaux , Infections à coronavirus/complications , Dipeptidyl peptidase 4/génétique , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Souris transgéniques , Fibrose pulmonaire/étiologie
8.
Int J Mol Sci ; 23(24)2022 Dec 10.
Article Dans Anglais | MEDLINE | ID: covidwho-2155138

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly contagious and pathogenic coronavirus that emerged in late 2019 and caused a pandemic of respiratory illness termed as coronavirus disease 2019 (COVID-19). Cancer patients are more susceptible to SARS-CoV-2 infection. The treatment of cancer patients infected with SARS-CoV-2 is more complicated, and the patients are at risk of poor prognosis compared to other populations. Patients infected with SARS-CoV-2 are prone to rapid development of acute respiratory distress syndrome (ARDS) of which pulmonary fibrosis (PF) is considered a sequelae. Both ARDS and PF are factors that contribute to poor prognosis in COVID-19 patients. However, the molecular mechanisms among COVID-19, ARDS and PF in COVID-19 patients with cancer are not well-understood. In this study, the common differentially expressed genes (DEGs) between COVID-19 patients with and without cancer were identified. Based on the common DEGs, a series of analyses were performed, including Gene Ontology (GO) and pathway analysis, protein-protein interaction (PPI) network construction and hub gene extraction, transcription factor (TF)-DEG regulatory network construction, TF-DEG-miRNA coregulatory network construction and drug molecule identification. The candidate drug molecules (e.g., Tamibarotene CTD 00002527) obtained by this study might be helpful for effective therapeutic targets in COVID-19 patients with cancer. In addition, the common DEGs among ARDS, PF and COVID-19 patients with and without cancer are TNFSF10 and IFITM2. These two genes may serve as potential therapeutic targets in the treatment of COVID-19 patients with cancer. Changes in the expression levels of TNFSF10 and IFITM2 in CD14+/CD16+ monocytes may affect the immune response of COVID-19 patients. Specifically, changes in the expression level of TNFSF10 in monocytes can be considered as an immune signature in COVID-19 patients with hematologic cancer. Targeting N6-methyladenosine (m6A) pathways (e.g., METTL3/SERPINA1 axis) to restrict SARS-CoV-2 reproduction has therapeutic potential for COVID-19 patients.


Sujets)
COVID-19 , Tumeurs , Fibrose pulmonaire , , Humains , COVID-19/complications , COVID-19/génétique , Poumon/anatomopathologie , Protéines membranaires/métabolisme , Methyltransferases/métabolisme , Tumeurs/complications , Tumeurs/génétique , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/virologie , /anatomopathologie , /virologie , RNA-Seq , SARS-CoV-2 , , Facteurs de transcription/métabolisme
9.
Biomolecules ; 12(12)2022 12 10.
Article Dans Anglais | MEDLINE | ID: covidwho-2154890

Résumé

The global scope and scale of the SARS-CoV-2 pandemic led to huge amounts of important data from clinical observations and experimental analyses being collected, in particular, regarding the long-term impact of COVID-19 on lung tissue. Visible changes in lung tissue mainly relate to the destruction of the alveolar architecture, dense cellularity, and pulmonary fibrosis with myofibroblast proliferation and collagen deposition. These changes are the result of infection, mainly with virus variants from the first pandemic waves (Alpha to Delta). In addition, proper regulation of immune responses to pathogenic viral stimuli is critical for the control of and recovery from tissue/organ damage, including in the lungs. We can distinguish three main processes in the lungs during SARS-CoV-2 infection: damage or deficiency of the pulmonary surfactant, coagulation processes, and fibrosis. Understanding the molecular basis of these processes is extremely important in the context of elucidating all pathologies occurring after virus entry. In the present review, data on the abovementioned three biochemical processes that lead to pathological changes are gathered together and discussed. Systematization of the knowledge is necessary to explore the three key pathways in lung tissue after SARS-CoV-2 virus infection as a result of a prolonged and intense inflammatory process in the context of pulmonary fibrosis, hemostatic disorders, and disturbances in the structure and/or metabolism of the surfactant. Despite the fact that the new Omicron variant does not affect the lungs as much as the previous variants, we cannot ignore the fact that other new mutations and emerging variants will not cause serious damage to the lung tissue. In the future, this review will be helpful to stratify the risk of serious complications in patients, to improve COVID-19 treatment outcomes, and to select those who may develop complications before clinical manifestation.


Sujets)
COVID-19 , Fibrose pulmonaire , Thrombose , Humains , COVID-19/génétique , COVID-19/anatomopathologie , SARS-CoV-2 , Fibrose pulmonaire/génétique , Fibrose pulmonaire/anatomopathologie , , Poumon/anatomopathologie , Thrombose/génétique , Thrombose/anatomopathologie
10.
Hum Genomics ; 16(1): 20, 2022 06 13.
Article Dans Anglais | MEDLINE | ID: covidwho-1951361

Résumé

The increased resolution of single-cell RNA-sequencing technologies has led to major breakthroughs and improved our understanding of the normal and pathologic conditions of multiple tissues and organs. In the study of parenchymal lung disease, single-cell RNA-sequencing has better delineated known cell populations and identified novel cells and changes in cellular phenotypes and gene expression patterns associated with disease. In this review, we aim to highlight the advances and insights that have been made possible by applying these technologies to two seemingly very different lung diseases: fibrotic interstitial lung diseases, a group of relentlessly progressive lung diseases leading to pulmonary fibrosis, and COVID-19 pneumonia, an acute viral disease with life-threatening complications, including pulmonary fibrosis. We discuss changes in cell populations and gene expression, highlighting potential common features, such as alveolar cell epithelial injury and aberrant repair and monocyte-derived macrophage populations, as well as relevance and implications to mechanisms of disease and future directions.


Sujets)
COVID-19 , Fibrose pulmonaire , COVID-19/génétique , Humains , Poumon/anatomopathologie , Fibrose pulmonaire/génétique , Fibrose pulmonaire/anatomopathologie , ARN , Analyse sur cellule unique
11.
Int J Mol Sci ; 23(3)2022 Jan 29.
Article Dans Anglais | MEDLINE | ID: covidwho-1667195

Résumé

(1) Background: In COVID-19 survivors there is an increased prevalence of pulmonary fibrosis of which the underlying molecular mechanisms are poorly understood; (2) Methods: In this multicentric study, n = 12 patients who succumbed to COVID-19 due to progressive respiratory failure were assigned to an early and late group (death within ≤7 and >7 days of hospitalization, respectively) and compared to n = 11 healthy controls; mRNA and protein expression as well as biological pathway analysis were performed to gain insights into the evolution of pulmonary fibrogenesis in COVID-19; (3) Results: Median duration of hospitalization until death was 3 (IQR25-75, 3-3.75) and 14 (12.5-14) days in the early and late group, respectively. Fifty-eight out of 770 analyzed genes showed a significantly altered expression signature in COVID-19 compared to controls in a time-dependent manner. The entire study group showed an increased expression of BST2 and IL1R1, independent of hospitalization time. In the early group there was increased activity of inflammation-related genes and pathways, while fibrosis-related genes (particularly PDGFRB) and pathways dominated in the late group; (4) Conclusions: After the first week of hospitalization, there is a shift from pro-inflammatory to fibrogenic activity in severe COVID-19. IL1R1 and PDGFRB may serve as potential therapeutic targets in future studies.


Sujets)
COVID-19/génétique , COVID-19/métabolisme , Fibrose pulmonaire/anatomopathologie , Sujet âgé , COVID-19/mortalité , Femelle , Mortalité hospitalière/tendances , Hospitalisation , Humains , Poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Fibrose pulmonaire/métabolisme , Insuffisance respiratoire/anatomopathologie , SARS-CoV-2/pathogénicité
12.
Int J Mol Sci ; 23(1)2021 Dec 24.
Article Dans Anglais | MEDLINE | ID: covidwho-1580700

Résumé

Acute respiratory distress syndrome (ARDS) followed by repair with lung remodeling is observed in COVID-19. These findings can lead to pulmonary terminal fibrosis, a form of irreversible sequelae. There is evidence that TGF-ß is intimately involved in the fibrogenic process. When activated, TGF-ß promotes the differentiation of fibroblasts into myofibroblasts and regulates the remodeling of the extracellular matrix (ECM). In this sense, the present study evaluated the histopathological features and immunohistochemical biomarkers (ACE-2, AKT-1, Caveolin-1, CD44v6, IL-4, MMP-9, α-SMA, Sphingosine-1, and TGF-ß1 tissue expression) involved in the TGF-ß1 signaling pathways and pulmonary fibrosis. The study consisted of 24 paraffin lung samples from patients who died of COVID-19 (COVID-19 group), compared to 10 lung samples from patients who died of H1N1pdm09 (H1N1 group) and 11 lung samples from patients who died of different causes, with no lung injury (CONTROL group). In addition to the presence of alveolar septal fibrosis, diffuse alveolar damage (DAD) was found to be significantly increased in the COVID-19 group, associated with a higher density of Collagen I (mature) and III (immature). There was also a significant increase observed in the immunoexpression of tissue biomarkers ACE-2, AKT-1, CD44v6, IL-4, MMP-9, α-SMA, Sphingosine-1, and TGF-ß1 in the COVID-19 group. A significantly lower expression of Caveolin-1 was also found in this group. The results suggest the participation of TGF-ß pathways in the development process of pulmonary fibrosis. Thus, it would be plausible to consider therapy with TGF-ß inhibitors in those patients recovered from COVID-19 to mitigate a possible development of pulmonary fibrosis and its consequences for post-COVID-19 life quality.


Sujets)
COVID-19/métabolisme , Fibrose pulmonaire/métabolisme , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Actines/métabolisme , Hormones corticosurrénaliennes/usage thérapeutique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Angiotensin-converting enzyme 2/métabolisme , COVID-19/complications , COVID-19/anatomopathologie , Cavéoline-1/métabolisme , Collagène de type I/métabolisme , Collagène de type III/métabolisme , Femelle , Humains , Antigènes CD44/métabolisme , Immunohistochimie , Sous-type H1N1 du virus de la grippe A/métabolisme , Grippe humaine/métabolisme , Grippe humaine/anatomopathologie , Interleukine-4/métabolisme , Mâle , Matrix metalloproteinase 9/métabolisme , Adulte d'âge moyen , Protéines proto-oncogènes c-akt/métabolisme , Fibrose pulmonaire/complications , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Études rétrospectives , Facteur de croissance transformant bêta-1/métabolisme ,
13.
Pan Afr Med J ; 40: 169, 2021.
Article Dans Anglais | MEDLINE | ID: covidwho-1566818

Résumé

Twenty months into the COVID-19 pandemic, we are still learning about the various long-term consequences of COVID-19 infection. While many patients do recover with minimal long-term consequences, some patients develop irreversible parenchymal and interstitial lung damage leading to diffuse pulmonary fibrosis. Unfortunately, these are some of the consequences of post-SARS-CoV-2 infection which thousands more people around the world will experience and which will outlast the pandemic for a long time to come. It is now being observed at various leading medical centres around the world that lung transplantation may be the only meaningful treatment available to a select group of patients experiencing serious lung damage and non-resolving COVID-19-associated respiratory failure, resulting from the triad of coronavirus infection, a hyper-inflammatory immune response to it and the inability of the human body to repair that injury.


Sujets)
COVID-19 , Transplantation pulmonaire , Fibrose pulmonaire , Humains , Incidence , Poumon/anatomopathologie , Pandémies , Fibrose pulmonaire/épidémiologie , Fibrose pulmonaire/étiologie , Fibrose pulmonaire/anatomopathologie , SARS-CoV-2
14.
Lancet Respir Med ; 9(5): 487-497, 2021 05.
Article Dans Anglais | MEDLINE | ID: covidwho-1537196

Résumé

BACKGROUND: Lung transplantation is a life-saving treatment for patients with end-stage lung disease; however, it is infrequently considered for patients with acute respiratory distress syndrome (ARDS) attributable to infectious causes. We aimed to describe the course of disease and early post-transplantation outcomes in critically ill patients with COVID-19 who failed to show lung recovery despite optimal medical management and were deemed to be at imminent risk of dying due to pulmonary complications. METHODS: We established a multi-institutional case series that included the first consecutive transplants for severe COVID-19-associated ARDS known to us in the USA, Italy, Austria, and India. De-identified data from participating centres-including information relating to patient demographics and pre-COVID-19 characteristics, pretransplantation disease course, perioperative challenges, pathology of explanted lungs, and post-transplantation outcomes-were collected by Northwestern University (Chicago, IL, USA) and analysed. FINDINGS: Between May 1 and Sept 30, 2020, 12 patients with COVID-19-associated ARDS underwent bilateral lung transplantation at six high-volume transplant centres in the USA (eight recipients at three centres), Italy (two recipients at one centre), Austria (one recipient), and India (one recipient). The median age of recipients was 48 years (IQR 41-51); three of the 12 patients were female. Chest imaging before transplantation showed severe lung damage that did not improve despite prolonged mechanical ventilation and extracorporeal membrane oxygenation. The lung transplant procedure was technically challenging, with severe pleural adhesions, hilar lymphadenopathy, and increased intraoperative transfusion requirements. Pathology of the explanted lungs showed extensive, ongoing acute lung injury with features of lung fibrosis. There was no recurrence of SARS-CoV-2 in the allografts. All patients with COVID-19 could be weaned off extracorporeal support and showed short-term survival similar to that of transplant recipients without COVID-19. INTERPRETATION: The findings from our report show that lung transplantation is the only option for survival in some patients with severe, unresolving COVID-19-associated ARDS, and that the procedure can be done successfully, with good early post-transplantation outcomes, in carefully selected patients. FUNDING: National Institutes of Health. VIDEO ABSTRACT.


Sujets)
COVID-19 , Maladie grave/thérapie , Transplantation pulmonaire/méthodes , Poumon , , Transfusion sanguine/méthodes , COVID-19/complications , COVID-19/diagnostic , COVID-19/physiopathologie , COVID-19/chirurgie , Soins de réanimation/méthodes , Oxygénation extracorporelle sur oxygénateur à membrane/méthodes , Femelle , Humains , Soins peropératoires/méthodes , Poumon/imagerie diagnostique , Poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Évaluation des résultats et des processus en soins de santé , Fibrose pulmonaire/étiologie , Fibrose pulmonaire/anatomopathologie , Ventilation artificielle/méthodes , /étiologie , /chirurgie , SARS-CoV-2/pathogénicité
15.
Front Immunol ; 12: 735922, 2021.
Article Dans Anglais | MEDLINE | ID: covidwho-1477823

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a major public health issue. COVID-19 is considered an airway/multi-systemic disease, and demise has been associated with an uncontrolled immune response and a cytokine storm in response to the virus. However, the lung pathology, immune response, and tissue damage associated with COVID-19 demise are poorly described and understood due to safety concerns. Using post-mortem lung tissues from uninfected and COVID-19 deadly cases as well as an unbiased combined analysis of histology, multi-viral and host markers staining, correlative microscopy, confocal, and image analysis, we identified three distinct phenotypes of COVID-19-induced lung damage. First, a COVID-19-induced hemorrhage characterized by minimal immune infiltration and large thrombus; Second, a COVID-19-induced immune infiltration with excessive immune cell infiltration but no hemorrhagic events. The third phenotype correspond to the combination of the two previous ones. We observed the loss of alveolar wall integrity, detachment of lung tissue pieces, fibroblast proliferation, and extensive fibrosis in all three phenotypes. Although lung tissues studied were from lethal COVID-19, a strong immune response was observed in all cases analyzed with significant B cell and poor T cell infiltrations, suggesting an exhausted or compromised immune cellular response in these patients. Overall, our data show that SARS-CoV-2-induced lung damage is highly heterogeneous. These individual differences need to be considered to understand the acute and long-term COVID-19 consequences.


Sujets)
COVID-19/mortalité , COVID-19/anatomopathologie , Lésion pulmonaire/anatomopathologie , Alvéoles pulmonaires/anatomopathologie , Fibrose pulmonaire/anatomopathologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Autopsie , Lymphocytes T CD8+/immunologie , Syndrome de libération de cytokines/mortalité , Syndrome de libération de cytokines/anatomopathologie , Cellules épithéliales/anatomopathologie , Femelle , Hémorragie/anatomopathologie , Humains , Inflammation/anatomopathologie , Poumon/anatomopathologie , Lésion pulmonaire/virologie , Lymphopénie/anatomopathologie , Activation des macrophages/immunologie , Macrophages/immunologie , Mâle , Adulte d'âge moyen , Myocytes du muscle lisse/anatomopathologie , Granulocytes neutrophiles/immunologie , SARS-CoV-2 , Thrombose/anatomopathologie
16.
Front Immunol ; 12: 687397, 2021.
Article Dans Anglais | MEDLINE | ID: covidwho-1477818

Résumé

Severe COVID-19 is characterized by acute respiratory distress syndrome (ARDS)-like hyperinflammation and endothelial dysfunction, that can lead to respiratory and multi organ failure and death. Interstitial lung diseases (ILD) and pulmonary fibrosis confer an increased risk for severe disease, while a subset of COVID-19-related ARDS surviving patients will develop a fibroproliferative response that can persist post hospitalization. Autotaxin (ATX) is a secreted lysophospholipase D, largely responsible for the extracellular production of lysophosphatidic acid (LPA), a pleiotropic signaling lysophospholipid with multiple effects in pulmonary and immune cells. In this review, we discuss the similarities of COVID-19, ARDS and ILDs, and suggest ATX as a possible pathologic link and a potential common therapeutic target.


Sujets)
COVID-19/anatomopathologie , Phosphodiesterases/métabolisme , Fibrose pulmonaire/anatomopathologie , /anatomopathologie , Anti-inflammatoires/usage thérapeutique , COVID-19/sang , Dexaméthasone/usage thérapeutique , Humains , Poumon/anatomopathologie , Lysophospholipides/métabolisme , Phosphodiesterases/sang , Fibrose pulmonaire/sang , /sang , SARS-CoV-2 , Transduction du signal/immunologie
17.
Sci Rep ; 11(1): 19979, 2021 10 07.
Article Dans Anglais | MEDLINE | ID: covidwho-1462032

Résumé

COVID-19 pandemic led to a worldwide increase of hospitalizations for interstitial pneumonia with thrombosis complications, endothelial injury and multiorgan disease. Common CT findings include lung bilateral infiltrates, bilateral ground-glass opacities and/or consolidation whilst no current laboratory parameter consents rapidly evaluation of COVID-19 risk and disease severity. In the present work we investigated the association of sFLT-1 and CA 15.3 with endothelial damage and pulmonary fibrosis. Serum sFlt-1 has been associated with endothelial injury and sepsis severity, CA 15.3 seems an alternative marker for KL-6 for fibrotic lung diseases and pulmonary interstitial damage. We analysed 262 SARS-CoV-2 patients with differing levels of clinical severity; we found an association of serum sFlt-1 (ROC AUC 0.902, decision threshold > 90.3 pg/mL, p < 0.001 Sens. 83.9% and Spec. 86.7%) with presence, extent and severity of the disease. Moreover, CA 15.3 appeared significantly increased in COVID-19 severe lung fibrosis (ICU vs NON-ICU patients 42.6 ± 3.3 vs 25.7 ± 1.5 U/mL, p < 0.0001) and was associated with lung damage severity grade (ROC AUC 0.958, decision threshold > 24.8 U/mL, p < 0.0001, Sens. 88.4% and Spec. 91.8%). In conclusion, serum levels of sFlt-1 and CA 15.3 appeared useful tools for categorizing COVID-19 clinical stage and may represent a valid aid for clinicians to better personalise treatment.


Sujets)
COVID-19/sang , Mucine-1/sang , Fibrose pulmonaire/sang , Récepteur-1 au facteur croissance endothéliale vasculaire/sang , Sujet âgé , Marqueurs biologiques/sang , COVID-19/complications , COVID-19/anatomopathologie , Femelle , Humains , Pneumopathies interstitielles/sang , Pneumopathies interstitielles/complications , Pneumopathies interstitielles/anatomopathologie , Mâle , Adulte d'âge moyen , Fibrose pulmonaire/complications , Fibrose pulmonaire/anatomopathologie , SARS-CoV-2/isolement et purification
18.
Adv Respir Med ; 89(5): 477-483, 2021.
Article Dans Anglais | MEDLINE | ID: covidwho-1456468

Résumé

INTRODUCTION: COVID-19-associated pulmonary sequalae have been increasingly reported after recovery from acute infection. Therefore, we aim to explore the charactersitics of persistent lung parenchymal abnormalities in patients with COVID-19. MATERIAL AND METHODS: An observational study was conducted in patients with post-COVID lung parenchymal abnormalities from April till September 2020. Patients ≥18 years of age with COVID-19 who were diagnosed as post-COVID lung parenchymal abnormality based on respiratory symptoms and HRCT chest imaging after the recovery of acute infection. Data was recorded on a structured pro forma, and descriptive analysis was performed using Stata version 12.1. RESULTS: A total of 30 patients with post-COVID lung parenchymal abnormalities were identified. The mean age of patients was 59.1 (SD 12.6), and 27 (90.0%) were males. Four HRCT patterns of lung parenchymal abnormalities were seen; organizing pneumonia in 10 (33.3%), nonspecific interstitial pneumonitis in 17 (56.7%), usual interstitial pneumonitis in 12 (40.0%) and probable usual interstitial pneumonitis in 14 (46.7%). Diffuse involvement was found in 15 (50.0%) patients, while peripheral predominance in 15 (50.0%), and other significant findings were seen in 8 (26.7%) patients. All individuals were treated with corticosteroids. The case fatality rate was 16.7%. Amongst the survivors, 32.0% recovered completely, 36.0% improved, while 32.0% of the patients had static or progressive disease. CONCLUSION: This is the first study from Southeast Asia that identified post-COVID lung parenchymal abnormalities in patients who had no pre-existing lung disease highlighting the importance of timely recognition and treatment of this entity that might lead to fatal outcome.


Sujets)
COVID-19/imagerie diagnostique , COVID-19/anatomopathologie , Fibrose pulmonaire/imagerie diagnostique , Fibrose pulmonaire/anatomopathologie , Adulte , COVID-19/complications , Femelle , Études de suivi , Humains , Mâle , Adulte d'âge moyen , Sortie du patient , SARS-CoV-2/isolement et purification , Tomodensitométrie
19.
J Ethnopharmacol ; 283: 114701, 2022 Jan 30.
Article Dans Anglais | MEDLINE | ID: covidwho-1446835

Résumé

ETHNOPHARMACOLOGICAL RELEVANCE: Xuanfei Baidu Decoction (XFBD), one of the "three medicines and three prescriptions" for the clinically effective treatment of COVID-19 in China, plays an important role in the treatment of mild and/or common patients with dampness-toxin obstructing lung syndrome. AIM OF THE STUDY: The present work aims to elucidate the protective effects and the possible mechanism of XFBD against the acute inflammation and pulmonary fibrosis. METHODS: We use TGF-ß1 induced fibroblast activation model and LPS/IL-4 induced macrophage inflammation model as in vitro cell models. The mice model of lung fibrosis was induced by BLM via endotracheal drip, and then XFBD (4.6 g/kg, 9.2 g/kg) were administered orally respectively. The efficacy and molecular mechanisms in the presence or absence of XFBD were investigated. RESULTS: The results proved that XFBD can effectively inhibit fibroblast collagen deposition, down-regulate the level of α-SMA and inhibit the migration of fibroblasts. IL-4 induced macrophage polarization was also inhibited and the secretions of the inflammatory factors including IL6, iNOS were down-regulated. In vivo experiments, the results proved that XFBD improved the weight loss and survival rate of the mice. The XFBD high-dose administration group had a significant effect in inhibiting collagen deposition and the expression of α-SMA in the lungs of mice. XFBD can reduce bleomycin-induced pulmonary fibrosis by inhibiting IL-6/STAT3 activation and related macrophage infiltration. CONCLUSIONS: Xuanfei Baidu Decoction protects against macrophages induced inflammation and pulmonary fibrosis via inhibiting IL-6/STAT3 signaling pathway.


Sujets)
, Médicaments issus de plantes chinoises , Inflammation/traitement médicamenteux , Macrophages/effets des médicaments et des substances chimiques , SARS-CoV-2 , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Fibroblastes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Réseaux de régulation génique , Humains , Interleukine-6/antagonistes et inhibiteurs , Interleukine-6/génétique , Interleukine-6/métabolisme , Mâle , Souris , Souris de lignée C57BL , Cellules NIH 3T3 , Phytothérapie , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/prévention et contrôle , Cellules RAW 264.7 , Facteur de transcription STAT-3/antagonistes et inhibiteurs , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/métabolisme
20.
Immunity ; 54(11): 2650-2669.e14, 2021 11 09.
Article Dans Anglais | MEDLINE | ID: covidwho-1442406

Résumé

Longitudinal analyses of the innate immune system, including the earliest time points, are essential to understand the immunopathogenesis and clinical course of coronavirus disease (COVID-19). Here, we performed a detailed characterization of natural killer (NK) cells in 205 patients (403 samples; days 2 to 41 after symptom onset) from four independent cohorts using single-cell transcriptomics and proteomics together with functional studies. We found elevated interferon (IFN)-α plasma levels in early severe COVD-19 alongside increased NK cell expression of IFN-stimulated genes (ISGs) and genes involved in IFN-α signaling, while upregulation of tumor necrosis factor (TNF)-induced genes was observed in moderate diseases. NK cells exert anti-SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) activity but are functionally impaired in severe COVID-19. Further, NK cell dysfunction may be relevant for the development of fibrotic lung disease in severe COVID-19, as NK cells exhibited impaired anti-fibrotic activity. Our study indicates preferential IFN-α and TNF responses in severe and moderate COVID-19, respectively, and associates a prolonged IFN-α-induced NK cell response with poorer disease outcome.


Sujets)
COVID-19/immunologie , Interféron alpha/immunologie , Cellules tueuses naturelles/immunologie , SARS-CoV-2/immunologie , Facteur de nécrose tumorale alpha/métabolisme , Séquence nucléotidique , Humains , Immunité innée/immunologie , Inflammation/immunologie , Interféron alpha/sang , Fibrose pulmonaire/anatomopathologie , RNA-Seq , Indice de gravité de la maladie , Transcriptome/génétique , Royaume-Uni , États-Unis
SÉLECTION CITATIONS
Détails de la recherche