Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 40
Filtre
1.
Int J Mol Sci ; 23(1)2021 Dec 23.
Article Dans Anglais | MEDLINE | ID: covidwho-1855640

Résumé

Macrophages are present in most human tissues and have very diverse functions. Activated macrophages are usually divided into two phenotypes, M1 macrophages and M2 macrophages, which are altered by various factors such as microorganisms, tissue microenvironment, and cytokine signals. Macrophage polarity is very important for infections, inflammatory diseases, and malignancies; its management can be key in the prevention and treatment of diseases. In this review, we assess the current state of knowledge on macrophage polarity and report on its prospects as a therapeutic target.


Sujets)
Polarité de la cellule/physiologie , Macrophages/anatomopathologie , Animaux , Cytokines/métabolisme , Maladie , Humains , Macrophages/métabolisme
2.
Nature ; 606(7914): 585-593, 2022 06.
Article Dans Anglais | MEDLINE | ID: covidwho-1815563

Résumé

Severe COVID-19 is characterized by persistent lung inflammation, inflammatory cytokine production, viral RNA and a sustained interferon (IFN) response, all of which are recapitulated and required for pathology in the SARS-CoV-2-infected MISTRG6-hACE2 humanized mouse model of COVID-19, which has a human immune system1-20. Blocking either viral replication with remdesivir21-23 or the downstream IFN-stimulated cascade with anti-IFNAR2 antibodies in vivo in the chronic stages of disease attenuates the overactive immune inflammatory response, especially inflammatory macrophages. Here we show that SARS-CoV-2 infection and replication in lung-resident human macrophages is a critical driver of disease. In response to infection mediated by CD16 and ACE2 receptors, human macrophages activate inflammasomes, release interleukin 1 (IL-1) and IL-18, and undergo pyroptosis, thereby contributing to the hyperinflammatory state of the lungs. Inflammasome activation and the accompanying inflammatory response are necessary for lung inflammation, as inhibition of the NLRP3 inflammasome pathway reverses chronic lung pathology. Notably, this blockade of inflammasome activation leads to the release of infectious virus by the infected macrophages. Thus, inflammasomes oppose host infection by SARS-CoV-2 through the production of inflammatory cytokines and suicide by pyroptosis to prevent a productive viral cycle.


Sujets)
COVID-19 , Inflammasomes , Macrophages , SARS-CoV-2 , Angiotensin-converting enzyme 2 , Animaux , COVID-19/anatomopathologie , COVID-19/physiopathologie , COVID-19/virologie , Humains , Inflammasomes/métabolisme , Interleukine-1 , Interleukine-18 , Poumon/anatomopathologie , Poumon/virologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Macrophages/virologie , Souris , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/virologie , Pyroptose , Récepteurs du fragment Fc des IgG , SARS-CoV-2/métabolisme , SARS-CoV-2/pathogénicité
3.
EBioMedicine ; 75: 103803, 2022 Jan.
Article Dans Anglais | MEDLINE | ID: covidwho-1587923

Résumé

BACKGROUND: The Coronavirus Disease 2019 (COVID-19) pandemic has been a great threat to global public health since 2020. Although the advance on vaccine development has been largely achieved, a strategy to alleviate immune overactivation in severe COVID-19 patients is still needed. The NLRP3 inflammasome is activated upon SARS-CoV-2 infection and associated with COVID-19 severity. However, the processes by which the NLRP3 inflammasome is involved in COVID-19 disease remain unclear. METHODS: We infected THP-1 derived macrophages, NLRP3 knockout mice, and human ACE2 transgenic mice with live SARS-CoV-2 in Biosafety Level 3 (BSL-3) laboratory. We performed quantitative real-time PCR for targeted viral or host genes from SARS-CoV-2 infected mouse tissues, conducted histological or immunofluorescence analysis in SARS-CoV-2 infected mouse tissues. We also injected intranasally AAV-hACE2 or intraperitoneally NLRP3 inflammasome inhibitor MCC950 before SARS-CoV-2 infection in mice as indicated. FINDINGS: We have provided multiple lines of evidence that the NLRP3 inflammasome plays an important role in the host immune response to SARS-CoV-2 invasion of the lungs. Inhibition of the NLRP3 inflammasome attenuated the release of COVID-19 related pro-inflammatory cytokines in cell cultures and mice. The severe pathology induced by SARS-CoV-2 in lung tissues was reduced in Nlrp3-/- mice compared to wild-type C57BL/6 mice. Finally, specific inhibition of the NLRP3 inflammasome by MCC950 alleviated excessive lung inflammation and thus COVID-19 like pathology in human ACE2 transgenic mice. INTERPRETATION: Inflammatory activation induced by SARS-CoV-2 is an important stimulator of COVID-19 related immunopathology. Targeting the NLRP3 inflammasome is a promising immune intervention against severe COVID-19 disease. FUNDING: This work was supported by grants from the Bureau of Frontier Sciences and Education, CAS (grant no. QYZDJ-SSW-SMC005 to Y.G.Y.), the key project of the CAS "Light of West China" Program (to D.Y.) and Yunnan Province (202001AS070023 to D.Y.).


Sujets)
COVID-19 , Poumon , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , SARS-CoV-2/immunologie , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Animaux , COVID-19/génétique , COVID-19/immunologie , COVID-19/anatomopathologie , Modèles animaux de maladie humaine , Humains , Poumon/immunologie , Poumon/anatomopathologie , Poumon/virologie , Macrophages/immunologie , Macrophages/anatomopathologie , Macrophages/virologie , Mâle , Souris , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , SARS-CoV-2/génétique , Cellules THP-1
4.
Cell Rep ; 37(12): 110126, 2021 12 21.
Article Dans Anglais | MEDLINE | ID: covidwho-1556413

Résumé

Previous studies have shown that the high mortality caused by viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus primarily results from complications of a cytokine storm. Therefore, it is critical to identify the key factors participating in the cytokine storm. Here we demonstrate that interferon-induced protein 35 (IFP35) plays an important role in the cytokine storm induced by SARS-CoV-2 and influenza virus infection. We find that the levels of serum IFP35 in individuals with SARS-CoV-2 correlates with severity of the syndrome. Using mouse model and cell assays, we show that IFP35 is released by lung epithelial cells and macrophages after SARS-CoV-2 or influenza virus infection. In addition, we show that administration of neutralizing antibodies against IFP35 considerably reduces lung injury and, thus, the mortality rate of mice exposed to viral infection. Our findings suggest that IFP35 serves as a biomarker and as a therapeutic target in virus-induced syndromes.


Sujets)
, COVID-19/sang , Grippe humaine/sang , Grippe humaine/traitement médicamenteux , Protéines et peptides de signalisation intracellulaire/sang , Animaux , Anticorps neutralisants/administration et posologie , Marqueurs biologiques/sang , COVID-19/anatomopathologie , COVID-19/physiopathologie , Modèles animaux de maladie humaine , Humains , Inflammation/métabolisme , Grippe humaine/anatomopathologie , Poumon/métabolisme , Poumon/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout , Acuité des besoins du patient , SARS-CoV-2/physiologie
5.
Cell ; 184(26): 6243-6261.e27, 2021 12 22.
Article Dans Anglais | MEDLINE | ID: covidwho-1536467

Résumé

COVID-19-induced "acute respiratory distress syndrome" (ARDS) is associated with prolonged respiratory failure and high mortality, but the mechanistic basis of lung injury remains incompletely understood. Here, we analyze pulmonary immune responses and lung pathology in two cohorts of patients with COVID-19 ARDS using functional single-cell genomics, immunohistology, and electron microscopy. We describe an accumulation of CD163-expressing monocyte-derived macrophages that acquired a profibrotic transcriptional phenotype during COVID-19 ARDS. Gene set enrichment and computational data integration revealed a significant similarity between COVID-19-associated macrophages and profibrotic macrophage populations identified in idiopathic pulmonary fibrosis. COVID-19 ARDS was associated with clinical, radiographic, histopathological, and ultrastructural hallmarks of pulmonary fibrosis. Exposure of human monocytes to SARS-CoV-2, but not influenza A virus or viral RNA analogs, was sufficient to induce a similar profibrotic phenotype in vitro. In conclusion, we demonstrate that SARS-CoV-2 triggers profibrotic macrophage responses and pronounced fibroproliferative ARDS.


Sujets)
COVID-19/anatomopathologie , COVID-19/virologie , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/virologie , Macrophages/anatomopathologie , Macrophages/virologie , SARS-CoV-2/physiologie , Antigènes CD/métabolisme , Antigènes de différenciation des myélomonocytes/métabolisme , COVID-19/imagerie diagnostique , Communication cellulaire , Études de cohortes , Fibroblastes/anatomopathologie , Régulation de l'expression des gènes , Humains , Fibrose pulmonaire idiopathique/imagerie diagnostique , Fibrose pulmonaire idiopathique/génétique , Cellules souches mésenchymateuses/anatomopathologie , Phénotype , Protéome/métabolisme , Récepteurs de surface cellulaire/métabolisme , /imagerie diagnostique , /anatomopathologie , /virologie , Tomodensitométrie , Transcription génétique
6.
Theranostics ; 12(1): 324-339, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-1512992

Résumé

Background: Macrophage infiltration around lipotoxic tubular epithelial cells (TECs) is a hallmark of diabetic nephropathy (DN). However, how these two types of cells communicate remains obscure. We previously demonstrated that LRG1 was elevated in the process of kidney injury. Here, we demonstrated that macrophage-derived, LRG1-enriched extracellular vesicles (EVs) exacerbated DN. Methods: We induced an experimental T2DM mouse model with a HFD diet for four months. Renal primary epithelial cells and macrophage-derived EVs were isolated from T2D mice by differential ultracentrifugation. To investigate whether lipotoxic TEC-derived EV (EVe) activate macrophages, mouse bone marrow-derived macrophages (BMDMs) were incubated with EVe. To investigate whether activated macrophage-derived EVs (EVm) induce lipotoxic TEC apoptosis, EVm were cocultured with primary renal tubular epithelial cells. Subsequently, we evaluated the effect of LRG1 in EVe by investigating the apoptosis mechanism. Results: We demonstrated that incubation of primary TECs of DN or HK-2 mTECs with lysophosphatidyl choline (LPC) increased the release of EVe. Interestingly, TEC-derived EVe activated an inflammatory phenotype in macrophages and induced the release of macrophage-derived EVm. Furthermore, EVm could induce apoptosis in TECs injured by LPC. Importantly, we found that leucine-rich α-2-glycoprotein 1 (LRG1)-enriched EVe activated macrophages via a TGFßR1-dependent process and that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-enriched EVm induced apoptosis in injured TECs via a death receptor 5 (DR5)-dependent process. Conclusion: Our findings indicated a novel cell communication mechanism between tubular epithelial cells and macrophages in DN, which could be a potential therapeutic target.


Sujets)
Diabète/métabolisme , Néphropathies diabétiques/métabolisme , Cellules épithéliales/métabolisme , Macrophages/métabolisme , Animaux , Apoptose , Communication cellulaire , Lignée cellulaire , Cellules épithéliales/anatomopathologie , Macrophages/anatomopathologie , Souris , Souris de lignée C57BL
7.
Arch Pathol Lab Med ; 145(11): 1328-1340, 2021 11 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1485410

Résumé

CONTEXT.­: SARS-CoV-2 can undergo maternal-fetal transmission, heightening interest in the placental pathology findings from this infection. Transplacental SARS-CoV-2 transmission is typically accompanied by chronic histiocytic intervillositis together with necrosis and positivity of syncytiotrophoblast for SARS-CoV-2. Hofbauer cells are placental macrophages that have been involved in viral diseases, including HIV and Zika virus, but their involvement in SARS-CoV-2 is unknown. OBJECTIVE.­: To determine whether SARS-CoV-2 can extend beyond the syncytiotrophoblast to enter Hofbauer cells, endothelium, and other villous stromal cells in infected placentas of liveborn and stillborn infants. DESIGN.­: Case-based retrospective analysis by 29 perinatal and molecular pathology specialists of placental findings from a preselected cohort of 22 SARS-CoV-2-infected placentas delivered to pregnant women testing positive for SARS-CoV-2 from 7 countries. Molecular pathology methods were used to investigate viral involvement of Hofbauer cells, villous capillary endothelium, syncytiotrophoblast, and other fetal-derived cells. RESULTS.­: Chronic histiocytic intervillositis and trophoblast necrosis were present in all 22 placentas (100%). SARS-CoV-2 was identified in Hofbauer cells from 4 of 22 placentas (18.2%). Villous capillary endothelial staining was positive in 2 of 22 cases (9.1%), both of which also had viral positivity in Hofbauer cells. Syncytiotrophoblast staining occurred in 21 of 22 placentas (95.5%). Hofbauer cell hyperplasia was present in 3 of 22 placentas (13.6%). In the 7 cases having documented transplacental infection of the fetus, 2 (28.6%) occurred in placentas with Hofbauer cell staining positive for SARS-CoV-2. CONCLUSIONS.­: SARS-CoV-2 can extend beyond the trophoblast into the villous stroma, involving Hofbauer cells and capillary endothelial cells, in a small number of infected placentas. Most cases of SARS-CoV-2 transplacental fetal infection occur without Hofbauer cell involvement.


Sujets)
COVID-19/transmission , COVID-19/virologie , Transmission verticale de maladie infectieuse , Macrophages/virologie , Placenta/virologie , Complications infectieuses de la grossesse/virologie , SARS-CoV-2/pathogénicité , Adulte , COVID-19/immunologie , COVID-19/anatomopathologie , Prolifération cellulaire , Endothélium/anatomopathologie , Endothélium/virologie , Femelle , Humains , Hyperplasie/anatomopathologie , Hyperplasie/virologie , Nouveau-né , Macrophages/anatomopathologie , Macrophages/physiologie , Mâle , Placenta/anatomopathologie , Grossesse , Complications infectieuses de la grossesse/immunologie , Complications infectieuses de la grossesse/anatomopathologie , Études rétrospectives , SARS-CoV-2/immunologie , Mortinatalité , Trophoblastes/anatomopathologie , Trophoblastes/virologie
8.
Cells ; 10(9)2021 08 31.
Article Dans Anglais | MEDLINE | ID: covidwho-1390541

Résumé

COVID-19 presents with a wide range of clinical neurological manifestations. It has been recognized that SARS-CoV-2 infection affects both the central and peripheral nervous system, leading to smell and taste disturbances; acute ischemic and hemorrhagic cerebrovascular disease; encephalopathies and seizures; and causes most surviving patients to have long lasting neurological symptoms. Despite this, typical neuropathological features associated with the infection have still not been identified. Studies of post-mortem examinations of the cerebral cortex are obtained with difficulty due to laboratory safety concerns. In addition, they represent cases with different neurological symptoms, age or comorbidities, thus a larger number of brain autoptic data from multiple institutions would be crucial. Histopathological findings described here are aimed to increase the current knowledge on neuropathology of COVID-19 patients. We report post-mortem neuropathological findings of ten COVID-19 patients. A wide range of neuropathological lesions were seen. The cerebral cortex of all patients showed vascular changes, hyperemia of the meninges and perivascular inflammation in the cerebral parenchyma with hypoxic neuronal injury. Perivascular lymphocytic inflammation of predominantly CD8-positive T cells mixed with CD68-positive macrophages, targeting the disrupted vascular wall in the cerebral cortex, cerebellum and pons were seen. Our findings support recent reports highlighting a role of microvascular injury in COVID-19 neurological manifestations.


Sujets)
COVID-19/anatomopathologie , Cortex cérébral/anatomopathologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Autopsie , Encéphale/anatomopathologie , Encéphale/virologie , Encéphalopathies/anatomopathologie , Encéphalopathies/virologie , Lymphocytes T CD8+/anatomopathologie , Cortex cérébral/virologie , Femelle , Humains , Inflammation , Macrophages/anatomopathologie , Mâle , Microvaisseaux/anatomopathologie , Microvaisseaux/virologie , Adulte d'âge moyen , Maladies du système nerveux/anatomopathologie , Maladies du système nerveux/virologie , SARS-CoV-2/pathogénicité
10.
Front Immunol ; 12: 720109, 2021.
Article Dans Anglais | MEDLINE | ID: covidwho-1348492

Résumé

COVID-19 is a contagious viral disease caused by SARS-CoV-2 that led to an ongoing pandemic with massive global health and socioeconomic consequences. The disease is characterized primarily, but not exclusively, by respiratory clinical manifestations ranging from mild common cold symptoms, including cough and fever, to severe respiratory distress and multi-organ failure. Macrophages, a heterogeneous group of yolk-sac derived, tissue-resident mononuclear phagocytes of complex ontogeny present in all mammalian organs, play critical roles in developmental, homeostatic and host defense processes with tissue-dependent plasticity. In case of infection, they are responsible for early pathogen recognition, initiation and resolution of inflammation, as well as repair of tissue damage. Monocytes, bone-marrow derived blood-resident phagocytes, are recruited under pathological conditions such as viral infections to the affected tissue to defend the organism against invading pathogens and to aid in efficient resolution of inflammation. Given their pivotal function in host defense and the potential danger posed by their dysregulated hyperinflammation, understanding monocyte and macrophage phenotypes in COVID-19 is key for tackling the disease's pathological mechanisms. Here, we outline current knowledge on monocytes and macrophages in homeostasis and viral infections and summarize concepts and key findings on their role in COVID-19. While monocytes in the blood of patients with moderate COVID-19 present with an inflammatory, interferon-stimulated gene (ISG)-driven phenotype, cellular dysfunction epitomized by loss of HLA-DR expression and induction of S100 alarmin expression is their dominant feature in severe disease. Pulmonary macrophages in COVID-19 derived from infiltrating inflammatory monocytes are in a hyperactivated state resulting in a detrimental loop of pro-inflammatory cytokine release and recruitment of cytotoxic effector cells thereby exacerbating tissue damage at the site of infection.


Sujets)
COVID-19/immunologie , Antigènes HLA-DR/immunologie , Macrophages/immunologie , Monocytes/immunologie , SARS-CoV-2/immunologie , COVID-19/anatomopathologie , Humains , Inflammation/immunologie , Inflammation/anatomopathologie , Macrophages/anatomopathologie , Monocytes/anatomopathologie , Indice de gravité de la maladie
11.
Cell Chem Biol ; 29(2): 239-248.e4, 2022 02 17.
Article Dans Anglais | MEDLINE | ID: covidwho-1347527

Résumé

Triggering receptor expressed on myeloid cells-2 (TREM2) is a cell surface receptor on macrophages and microglia that senses and responds to disease-associated signals to regulate the phenotype of these innate immune cells. The TREM2 signaling pathway has been implicated in a variety of diseases ranging from neurodegeneration in the central nervous system to metabolic disease in the periphery. Here, we report that TREM2 is a thyroid hormone-regulated gene and its expression in macrophages and microglia is stimulated by thyroid hormone and synthetic thyroid hormone agonists (thyromimetics). Our findings report the endocrine regulation of TREM2 by thyroid hormone, and provide a unique opportunity to drug the TREM2 signaling pathway with orally active small-molecule therapeutic agents.


Sujets)
Acétates/pharmacologie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Glycoprotéines membranaires/génétique , Microglie/effets des médicaments et des substances chimiques , Phénols/pharmacologie , Récepteurs immunologiques/génétique , Récepteurs X des rétinoïdes/génétique , Hormones thyroïdiennes/pharmacologie , Acétates/synthèse chimique , Animaux , Sites de fixation , Encéphale/effets des médicaments et des substances chimiques , Encéphale/immunologie , Encéphale/anatomopathologie , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Régulation de l'expression des gènes , Humains , Immunité innée , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/anatomopathologie , Glycoprotéines membranaires/antagonistes et inhibiteurs , Glycoprotéines membranaires/immunologie , Souris , Souris de lignée C57BL , Microglie/immunologie , Microglie/anatomopathologie , Modèles moléculaires , Phénols/synthèse chimique , Phénoxy-acétates/pharmacologie , Régions promotrices (génétique) , Liaison aux protéines , Structure en hélice alpha , Structure en brin bêta , Motifs et domaines d'intéraction protéique , ARN messager/antagonistes et inhibiteurs , ARN messager/génétique , ARN messager/immunologie , Récepteurs immunologiques/antagonistes et inhibiteurs , Récepteurs immunologiques/immunologie , Éléments de réponse , Récepteurs X des rétinoïdes/composition chimique , Récepteurs X des rétinoïdes/métabolisme , Transduction du signal
12.
Cell Commun Signal ; 19(1): 73, 2021 07 08.
Article Dans Anglais | MEDLINE | ID: covidwho-1301855

Résumé

BACKGROUND: The coronavirus disease 2019 (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) has become an ongoing pandemic. Understanding the respiratory immune microenvironment which is composed of multiple cell types, together with cell communication based on ligand-receptor interactions is important for developing vaccines, probing COVID-19 pathogenesis, and improving pandemic control measures. METHODS: A total of 102 consecutive hospitalized patients with confirmed COVID-19 were enrolled in this study. Clinical information, routine laboratory tests, and flow cytometry analysis data with different conditions were collected and assessed for predictive value in COVID-19 patients. Next, we analyzed public single-cell RNA-sequencing (scRNA-seq) data from bronchoalveolar lavage fluid, which offers the closest available view of immune cell heterogeneity as encountered in patients with varying severity of COVID-19. A weighting algorithm was used to calculate ligand-receptor interactions, revealing the communication potentially associated with outcomes across cell types. Finally, serum cytokines including IL6, IL1ß, IL10, CXCL10, TNFα, GALECTIN-1, and IGF1 derived from patients were measured. RESULTS: Of the 102 COVID-19 patients, 42 cases (41.2%) were categorized as severe. Multivariate logistic regression analysis demonstrated that AST, D-dimer, BUN, and WBC were considered as independent risk factors for the severity of COVID-19. T cell numbers including total T cells, CD4+ and CD8+ T cells in the severe disease group were significantly lower than those in the moderate disease group. The risk model containing the above mentioned inflammatory damage parameters, and the counts of T cells, with AUROCs ranged from 0.78 to 0.87. To investigate the molecular mechanism at the cellular level, we analyzed the published scRNA-seq data and found that macrophages displayed specific functional diversity after SARS-Cov-2 infection, and the metabolic pathway activities in the identified macrophage subtypes were influenced by hypoxia status. Importantly, we described ligand-receptor interactions that are related to COVID-19 serverity involving macrophages and T cell subsets by communication analysis. CONCLUSIONS: Our study showed that macrophages driving ligand-receptor crosstalk contributed to the reduction and exhaustion of CD8+ T cells. The identified crucial cytokine panel, including IL6, IL1ß, IL10, CXCL10, IGF1, and GALECTIN-1, may offer the selective targets to improve the efficacy of COVID-19 therapy. TRIAL REGISTRATION: This is a retrospective observational study without a trial registration number. Video Abstract.


Sujets)
COVID-19/immunologie , COVID-19/anatomopathologie , Communication cellulaire , Macrophages/immunologie , Analyse sur cellule unique , Sujet âgé , Liquide de lavage bronchoalvéolaire/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , COVID-19/épidémiologie , COVID-19/physiopathologie , Chine/épidémiologie , Cytokines/sang , Cytokines/immunologie , Femelle , Humains , Macrophages/anatomopathologie , Mâle , Adulte d'âge moyen , Récepteurs aux cytokines , Études rétrospectives , Analyse de séquence d'ARN , Indice de gravité de la maladie
13.
JAMA Neurol ; 78(8): 948-960, 2021 08 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1265359

Résumé

Importance: Myalgia, increased levels of creatine kinase, and persistent muscle weakness have been reported in patients with COVID-19. Objective: To study skeletal muscle and myocardial inflammation in patients with COVID-19 who had died. Design, Setting, and Participants: This case-control autopsy series was conducted in a university hospital as a multidisciplinary postmortem investigation. Patients with COVID-19 or other critical illnesses who had died between March 2020 and February 2021 and on whom an autopsy was performed were included. Individuals for whom informed consent to autopsy was available and the postmortem interval was less than 6 days were randomly selected. Individuals who were infected with SARS-CoV-2 per polymerase chain reaction test results and had clinical features suggestive of COVID-19 were compared with individuals with negative SARS-CoV-2 polymerase chain reaction test results and an absence of clinical features suggestive of COVID-19. Main Outcomes and Measures: Inflammation of skeletal muscle tissue was assessed by quantification of immune cell infiltrates, expression of major histocompatibility complex (MHC) class I and class II antigens on the sarcolemma, and a blinded evaluation on a visual analog scale ranging from absence of pathology to the most pronounced pathology. Inflammation of cardiac muscles was assessed by quantification of immune cell infiltrates. Results: Forty-three patients with COVID-19 (median [interquartile range] age, 72 [16] years; 31 men [72%]) and 11 patients with diseases other than COVID-19 (median [interquartile range] age, 71 [5] years; 7 men [64%]) were included. Skeletal muscle samples from the patients who died with COVID-19 showed a higher overall pathology score (mean [SD], 3.4 [1.8] vs 1.5 [1.0]; 95% CI, 0-3; P < .001) and a higher inflammation score (mean [SD], 3.5 [2.1] vs 1.0 [0.6]; 95% CI, 0-4; P < .001). Relevant expression of MHC class I antigens on the sarcolemma was present in 23 of 42 specimens from patients with COVID-19 (55%) and upregulation of MHC class II antigens in 7 of 42 specimens from patients with COVID-19 (17%), but neither were found in any of the controls. Increased numbers of natural killer cells (median [interquartile range], 8 [8] vs 3 [4] cells per 10 high-power fields; 95% CI, 1-10 cells per 10 high-power fields; P < .001) were found. Skeletal muscles showed more inflammatory features than cardiac muscles, and inflammation was most pronounced in patients with COVID-19 with chronic courses. In some muscle specimens, SARS-CoV-2 RNA was detected by reverse transcription-polymerase chain reaction, but no evidence for a direct viral infection of myofibers was found by immunohistochemistry and electron microscopy. Conclusions and Relevance: In this case-control study of patients who had died with and without COVID-19, most individuals with severe COVID-19 showed signs of myositis ranging from mild to severe. Inflammation of skeletal muscles was associated with the duration of illness and was more pronounced than cardiac inflammation. Detection of viral load was low or negative in most skeletal and cardiac muscles and probably attributable to circulating viral RNA rather than genuine infection of myocytes. This suggests that SARS-CoV-2 may be associated with a postinfectious, immune-mediated myopathy.


Sujets)
COVID-19/anatomopathologie , Muscles squelettiques/anatomopathologie , Myocardite/anatomopathologie , Myocarde/anatomopathologie , Myosite/anatomopathologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Autopsie , Lymphocytes T CD8+/anatomopathologie , COVID-19/métabolisme , Détection de l'acide nucléique du virus de la COVID-19 , Dépistage sérologique de la COVID-19 , Études cas-témoins , Femelle , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe II/métabolisme , Humains , Cellules tueuses naturelles/anatomopathologie , Leucocytes/anatomopathologie , Macrophages/anatomopathologie , Mâle , Adulte d'âge moyen , Muscles squelettiques/métabolisme , Myocardite/métabolisme , Myocarde/métabolisme , Myosite/métabolisme , ARN viral/métabolisme , SARS-CoV-2 , Sarcolemme/métabolisme , Facteurs temps
14.
Scand J Clin Lab Invest ; 81(4): 255-263, 2021 07.
Article Dans Anglais | MEDLINE | ID: covidwho-1242057

Résumé

Coronaviruses belonging to the Coronaviridae family are single-stranded RNA viruses. The entry of SARS-CoV-2 is accomplished via ACE-2 receptors. SARS-CoV-2 infection coactivates both innate and adaptive immune responses. Although SARS-CoV-2 stimulates antibody production with a typical pattern of IgM/IgG, cellular immunity is also impaired. In severe cases, low CD4 + and CD8 + T cell counts are associated with impaired immune functions, and high neutrophil/lymphocyte ratios accompanying low lymphocyte subsets have been demonstrated. Recently, high IFN -α/γ ratios with impaired T cell responses, and increased IL-1, IL-6, TNF-α, MCP-1, IP-10, IL-4, IL-10 have been reported in COVID-19 infection. Increased proinflammatory cytokines and chemokines in patients with severe COVID-19 may cause the suppression of CD4 + and CD8 + T cells and regulatory T cells, causing excessive inflammatory responses and fatal cytokine storm with tissue and organ damage. Consequently, novel therapeutics to be developed against host immune system, including blockade of cytokines (IL-6, IL-1, IFN) themselves, their receptors or signaling pathways- JAK inhibitors- could be effective as potential therapeutics.


Sujets)
Antiviraux/pharmacologie , , COVID-19/immunologie , COVID-19/physiopathologie , Hormones corticosurrénaliennes/usage thérapeutique , Animaux , Antiviraux/usage thérapeutique , Syndrome de libération de cytokines/traitement médicamenteux , Syndrome de libération de cytokines/étiologie , Syndrome de libération de cytokines/virologie , Cytokines/antagonistes et inhibiteurs , Cytokines/métabolisme , Glucocorticoïdes/usage thérapeutique , Humains , Hydroxychloroquine/usage thérapeutique , Immunothérapie/méthodes , Macrophages/immunologie , Macrophages/anatomopathologie , Macrophages/virologie
15.
Ann Diagn Pathol ; 53: 151744, 2021 Aug.
Article Dans Anglais | MEDLINE | ID: covidwho-1227970

Résumé

OBJECTIVES: Assess the pathologic changes in the lungs of COVID-19 decedents and correlate these changes with demographic data, clinical course, therapies, and duration of illness. METHODS: Lungs of 12 consecutive COVID-19 decedents consented for autopsy were evaluated for gross and histopathologic abnormalities. A complete Ghon "en block" dissection was performed on all cases; lung weights and gross characteristics recorded. Immunohistochemical studies were performed to characterize lymphocytic infiltrates and to assess SARS-CoV-2 capsid protein. RESULTS: Two distinct patterns of pulmonary involvement were identified. Three of 12 cases demonstrated a predominance of acute alveolar damage (DAD) while 9 of 12 cases demonstrated a marked increase in intra-alveolar macrophages in a fashion resembling desquamative interstitial pneumonia or macrophage activation syndrome (DIP/MAS). Two patterns were correlated solely with a statistically significant difference in the duration of illness. The group exhibiting DAD had duration of illness of 5.7 days while the group with DIP/MAS had duration of illness of 21.5 days (t-test p = 0.014). CONCLUSIONS: The pulmonary pathology of COVID-19 patients demonstrates a biphasic pattern, an acute phase demonstrating DAD changes while the patients with a more prolonged course exhibit a different pattern that resembles DIP/MAS-like pattern. The potential mechanisms and clinical significance are discussed.


Sujets)
COVID-19/anatomopathologie , Immunohistochimie/méthodes , Pneumopathies interstitielles/anatomopathologie , Poumon/anatomopathologie , Syndrome d'activation macrophagique/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Autopsie , COVID-19/complications , COVID-19/diagnostic , COVID-19/virologie , Protéines de capside/métabolisme , Comorbidité , Femelle , Humains , Poumon/métabolisme , Pneumopathies interstitielles/étiologie , Pneumopathies interstitielles/virologie , Lymphocytes/métabolisme , Lymphocytes/anatomopathologie , Syndrome d'activation macrophagique/étiologie , Syndrome d'activation macrophagique/virologie , Macrophages/anatomopathologie , Mâle , Adulte d'âge moyen , Alvéoles pulmonaires/immunologie , Alvéoles pulmonaires/anatomopathologie , SARS-CoV-2/génétique , Congé maladie
16.
Nature ; 595(7865): 114-119, 2021 07.
Article Dans Anglais | MEDLINE | ID: covidwho-1207147

Résumé

Respiratory failure is the leading cause of death in patients with severe SARS-CoV-2 infection1,2, but the host response at the lung tissue level is poorly understood. Here we performed single-nucleus RNA sequencing of about 116,000 nuclei from the lungs of nineteen individuals who died of COVID-19 and underwent rapid autopsy and seven control individuals. Integrated analyses identified substantial alterations in cellular composition, transcriptional cell states, and cell-to-cell interactions, thereby providing insight into the biology of lethal COVID-19. The lungs from individuals with COVID-19 were highly inflamed, with dense infiltration of aberrantly activated monocyte-derived macrophages and alveolar macrophages, but had impaired T cell responses. Monocyte/macrophage-derived interleukin-1ß and epithelial cell-derived interleukin-6 were unique features of SARS-CoV-2 infection compared to other viral and bacterial causes of pneumonia. Alveolar type 2 cells adopted an inflammation-associated transient progenitor cell state and failed to undergo full transition into alveolar type 1 cells, resulting in impaired lung regeneration. Furthermore, we identified expansion of recently described CTHRC1+ pathological fibroblasts3 contributing to rapidly ensuing pulmonary fibrosis in COVID-19. Inference of protein activity and ligand-receptor interactions identified putative drug targets to disrupt deleterious circuits. This atlas enables the dissection of lethal COVID-19, may inform our understanding of long-term complications of COVID-19 survivors, and provides an important resource for therapeutic development.


Sujets)
COVID-19/anatomopathologie , COVID-19/virologie , Poumon/anatomopathologie , SARS-CoV-2/pathogénicité , Analyse sur cellule unique , Sujet âgé , Sujet âgé de 80 ans ou plus , Pneumocytes/anatomopathologie , Pneumocytes/virologie , Atlas comme sujet , Autopsie , COVID-19/immunologie , Études cas-témoins , Femelle , Fibroblastes/anatomopathologie , Fibrose/anatomopathologie , Fibrose/virologie , Humains , Inflammation/anatomopathologie , Inflammation/virologie , Macrophages/anatomopathologie , Macrophages/virologie , Macrophages alvéolaires/anatomopathologie , Macrophages alvéolaires/virologie , Mâle , Adulte d'âge moyen , Plasmocytes/immunologie , Lymphocytes T/immunologie
17.
Cells ; 10(4)2021 04 14.
Article Dans Anglais | MEDLINE | ID: covidwho-1186898

Résumé

Macrophages play an important role in the innate and adaptive immune responses of organ systems, including the lungs, to particles and pathogens. Cumulative results show that macrophages contribute to the development and progression of acute or chronic inflammatory responses through the secretion of inflammatory cytokines/chemokines and the activation of transcription factors in the pathogenesis of inflammatory lung diseases, such as acute lung injury (ALI), acute respiratory distress syndrome (ARDS), ARDS related to COVID-19 (coronavirus disease 2019, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)), allergic asthma, chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). This review summarizes the functions of macrophages and their associated underlying mechanisms in the development of ALI, ARDS, COVID-19-related ARDS, allergic asthma, COPD, and IPF and briefly introduces the acute and chronic experimental animal models. Thus, this review suggests an effective therapeutic approach that focuses on the regulation of macrophage function in the context of inflammatory lung diseases.


Sujets)
Lésion pulmonaire aigüe/anatomopathologie , COVID-19/anatomopathologie , Macrophages/anatomopathologie , Pneumopathie infectieuse/anatomopathologie , Broncho-pneumopathie chronique obstructive/anatomopathologie , /anatomopathologie , Lésion pulmonaire aigüe/immunologie , Animaux , Asthme/immunologie , Asthme/anatomopathologie , COVID-19/immunologie , Maladie chronique , Humains , Fibrose pulmonaire idiopathique/immunologie , Fibrose pulmonaire idiopathique/anatomopathologie , Poumon/immunologie , Poumon/anatomopathologie , Macrophages/immunologie , Pneumopathie infectieuse/immunologie , Broncho-pneumopathie chronique obstructive/immunologie , /immunologie , SARS-CoV-2/immunologie
19.
Life Sci ; 277: 119503, 2021 Jul 15.
Article Dans Anglais | MEDLINE | ID: covidwho-1185151

Résumé

Severe coronavirus disease 2019 (COVID-19) caused by the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) is characterized by an unpredictable disease course, with variable presentations of different organ systems. The clinical manifestations of COVID-19 are highly variable ranging from mild presentations to severe, life-threatening symptoms and the wide individual variability may be due to the broad heterogeneity in the underlying pathologies. There is no doubt that early management may have a major influence on the outcome. This led the scientists to search for ways to monitor disease progression or to predict outcomes in COVID-19. Although it is not yet possible to predict who will progress to the severe forms or in what time, numerous prospective and longitudinal studies represent the evidence for determining the potential immunological risk factors of COVID-19 critical disease and death. The kinetics and breadth of immune responses during COVID-19 appear to follow a trend which is consistent to the predominant pathological alterations. Recent publications have used these biomarkers to help identify patients who will develop the severe acute COVID-19. Of particular interest is the relationship between the kinetics of peripheral leukocytes and clinical progress of the disease in COVID-19. Although research is ongoing in this area, we present details about the current status of the evaluation. Understanding of the COVID-19 related alterations of the innate and adaptive immune responses may help to promote the vaccine development and immunological interventions.


Sujets)
COVID-19/immunologie , Leucocytes/immunologie , SARS-CoV-2/immunologie , COVID-19/étiologie , COVID-19/anatomopathologie , COVID-19/thérapie , Évolution de la maladie , Humains , Immunité cellulaire , Immunité innée , Immunothérapie , Numération des leucocytes , Leucocytes/anatomopathologie , Macrophages/immunologie , Macrophages/anatomopathologie , Facteurs de risque , SARS-CoV-2/isolement et purification , Lymphocytes T/immunologie , Lymphocytes T/anatomopathologie
20.
Med Sci Monit ; 27: e928837, 2021 Feb 13.
Article Dans Anglais | MEDLINE | ID: covidwho-1161104

Résumé

BACKGROUND Coronavirus 2 (SARS-CoV-2) was declared a pandemic by the World Health Organization (WHO) in March 2020. To further reveal the pathologic associations between coronavirus and hypoxemia, we report the findings of 4 complete systematic autopsies of severe acute respiratory syndrome coronavirus 2-positive individuals who died of multiple organ failure caused by severe hypoxemia. MATERIAL AND METHODS We examined the donated corpses of 4 deceased patients who had been diagnosed with severe acute respiratory syndrome coronavirus 2. A complete post-mortem examination was carried out on each corpse, and multiple organs were macroscopically examined. RESULTS The 4 corpses were 2 males and 2 females, with an average age of 69 years. Bilateral lungs showed various degrees of atrophy and consolidation, with diffusely tough and solid texture in the sections. A thromboembolism was found in the main pulmonary artery extending into the atrium in 1 corpse, and significant atherosclerotic plaques tagged in the inner wall of the aortic arch were found in 2 corpses. Two corpses were found to have slightly atrophied bilateral renal parenchyma. Atrophic changes in the spleen were found in 2 corpses. Notably, there were significantly expanded alveolar septa and prominent fibroblastic proliferation. CONCLUSIONS The laboratory data of these corpses showed a progressive decrease in blood oxygen saturation, followed by refractory and irreversible hypoxemia. Clinical and laboratory information and autopsy and histologic presentations of multiple organs showed insufficient air exchange due to abnormalities in the respiratory system, and reduced erythropoiesis in bone marrow may play a role.


Sujets)
Autopsie , COVID-19/anatomopathologie , COVID-19/virologie , Hypoxie/complications , Hypoxie/anatomopathologie , Pneumopathie infectieuse/anatomopathologie , Pneumopathie infectieuse/virologie , SARS-CoV-2/physiologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Pneumocytes/métabolisme , Pneumocytes/anatomopathologie , COVID-19/complications , Agrégation cellulaire , Femelle , Humains , Poumon/anatomopathologie , Macrophages/anatomopathologie , Mâle , Adulte d'âge moyen , Mucus/métabolisme , Myocarde/anatomopathologie , Nécrose , Pneumopathie infectieuse/complications , Cavité thoracique/anatomopathologie
SÉLECTION CITATIONS
Détails de la recherche