Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtre
1.
Front Immunol ; 13: 820131, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-1731776

Résumé

Coronavirus disease 2019 (COVID-19) is currently a worldwide emergency caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). In observational clinical studies, statins have been identified as beneficial to hospitalized patients with COVID-19. However, experimental evidence of underlying statins protection against SARS-CoV-2 remains elusive. Here we reported for the first-time experimental evidence of the protective effects of simvastatin treatment both in vitro and in vivo. We found that treatment with simvastatin significantly reduced the viral replication and lung damage in vivo, delaying SARS-CoV-2-associated physiopathology and mortality in the K18-hACE2-transgenic mice model. Moreover, simvastatin also downregulated the inflammation triggered by SARS-CoV-2 infection in pulmonary tissue and in human neutrophils, peripheral blood monocytes, and lung epithelial Calu-3 cells in vitro, showing its potential to modulate the inflammatory response both at the site of infection and systemically. Additionally, we also observed that simvastatin affected the course of SARS-CoV-2 infection through displacing ACE2 on cell membrane lipid rafts. In conclusion, our results show that simvastatin exhibits early protective effects on SARS-CoV-2 infection by inhibiting virus cell entry and inflammatory cytokine production, through mechanisms at least in part dependent on lipid rafts disruption.


Sujets)
, Régulation négative/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Microdomaines membranaires/effets des médicaments et des substances chimiques , SARS-CoV-2/pathogénicité , Simvastatine/pharmacologie , Animaux , COVID-19/virologie , Modèles animaux de maladie humaine , Humains , Inflammation/virologie , Poumon/virologie , Souris , Souris transgéniques , Réplication virale/effets des médicaments et des substances chimiques
2.
Cell Metab ; 34(3): 424-440.e7, 2022 03 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1676683

Résumé

Coronavirus disease 2019 (COVID-19) represents a systemic disease that may cause severe metabolic complications in multiple tissues including liver, kidney, and cardiovascular system. However, the underlying mechanisms and optimal treatment remain elusive. Our study shows that impairment of ACE2 pathway is a key factor linking virus infection to its secondary metabolic sequelae. By using structure-based high-throughput virtual screening and connectivity map database, followed with experimental validations, we identify imatinib, methazolamide, and harpagoside as direct enzymatic activators of ACE2. Imatinib and methazolamide remarkably improve metabolic perturbations in vivo in an ACE2-dependent manner under the insulin-resistant state and SARS-CoV-2-infected state. Moreover, viral entry is directly inhibited by these three compounds due to allosteric inhibition of ACE2 binding to spike protein on SARS-CoV-2. Taken together, our study shows that enzymatic activation of ACE2 via imatinib, methazolamide, or harpagoside may be a conceptually new strategy to treat metabolic sequelae of COVID-19.


Sujets)
, Mésilate d'imatinib/usage thérapeutique , Maladies métaboliques/traitement médicamenteux , Méthazolamide/usage thérapeutique , SARS-CoV-2/effets des médicaments et des substances chimiques , Angiotensin-converting enzyme 2/effets des médicaments et des substances chimiques , Angiotensin-converting enzyme 2/métabolisme , Animaux , COVID-19/complications , COVID-19/métabolisme , COVID-19/virologie , Cellules cultivées , Chlorocebus aethiops , Régulation négative/effets des médicaments et des substances chimiques , Cellules HEK293 , Cellules endothéliales de la veine ombilicale humaine , Humains , Mésilate d'imatinib/pharmacologie , Mâle , Maladies métaboliques/métabolisme , Maladies métaboliques/virologie , Méthazolamide/pharmacologie , Souris , Souris de lignée C57BL , Souris obèse , Souris transgéniques , SARS-CoV-2/physiologie , Cellules Vero , Pénétration virale/effets des médicaments et des substances chimiques
3.
PLoS One ; 17(1): e0262737, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-1631070

Résumé

INTRODUCTION: The coronavirus disease 2019 (COVID-19), emerged in late 2019, was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The risk factors for idiopathic pulmonary fibrosis (IPF) and COVID-19 are reported to be common. This study aimed to determine the potential role of differentially expressed genes (DEGs) common in IPF and COVID-19. MATERIALS AND METHODS: Based on GEO database, we obtained DEGs from one SARS-CoV-2 dataset and five IPF datasets. A series of enrichment analysis were performed to identify the function of upregulated and downregulated DEGs, respectively. Two plugins in Cytoscape, Cytohubba and MCODE, were utilized to identify hub genes after a protein-protein interaction (PPI) network. Finally, candidate drugs were predicted to target the upregulated DEGs. RESULTS: A total of 188 DEGs were found between COVID-19 and IPF, out of which 117 were upregulated and 71 were downregulated. The upregulated DEGs were involved in cytokine function, while downregulated DEGs were associated with extracellular matrix disassembly. Twenty-two hub genes were upregulated in COVID-19 and IPF, for which 155 candidate drugs were predicted (adj.P.value < 0.01). CONCLUSION: Identifying the hub genes aberrantly regulated in both COVID-19 and IPF may enable development of molecules, encoded by those genes, as therapeutic targets for preventing IPF progression and SARS-CoV-2 infections.


Sujets)
COVID-19/génétique , Fibrose pulmonaire idiopathique/génétique , COVID-19/anatomopathologie , COVID-19/virologie , Bases de données génétiques , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Humains , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/anatomopathologie , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Cartes d'interactions protéiques/génétique , SARS-CoV-2/isolement et purification , Suloctidil/pharmacologie , Suloctidil/usage thérapeutique , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/génétique , Vasodilatateurs/pharmacologie , Vasodilatateurs/usage thérapeutique
4.
Sci Rep ; 11(1): 24432, 2021 12 24.
Article Dans Anglais | MEDLINE | ID: covidwho-1585772

Résumé

Despite the initial success of some drugs and vaccines targeting COVID-19, understanding the mechanism underlying SARS-CoV-2 disease pathogenesis remains crucial for the development of further approaches to treatment. Some patients with severe Covid-19 experience a cytokine storm and display evidence of inflammasome activation leading to increased levels of IL-1ß and IL-18; however, other reports have suggested reduced inflammatory responses to Sars-Cov-2. In this study we have examined the effects of the Sars-Cov-2 envelope (E) protein, a virulence factor in coronaviruses, on inflammasome activation and pulmonary inflammation. In cultured macrophages the E protein suppressed inflammasome priming and NLRP3 inflammasome activation. Similarly, in mice transfected with E protein and treated with poly(I:C) to simulate the effects of viral RNA, the E protein, in an NLRP3-dependent fashion, reduced expression of pro-IL-1ß, levels of IL-1ß and IL-18 in broncho-alveolar lavage fluid, and macrophage infiltration in the lung. To simulate the effects of more advanced infection, macrophages were treated with both LPS and poly(I:C). In this setting the E protein increased NLRP3 inflammasome activation in both murine and human macrophages. Thus, the Sars-Cov-2 E protein may initially suppress the host NLRP3 inflammasome response to viral RNA while potentially increasing NLRP3 inflammasome responses in the later stages of infection. Targeting the Sars-Cov-2 E protein especially in the early stages of infection may represent a novel approach to Covid-19 therapy.


Sujets)
Protéines d'enveloppe des coronavirus/métabolisme , Inflammasomes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , SARS-CoV-2/métabolisme , Animaux , Liquide de lavage bronchoalvéolaire/composition chimique , COVID-19/anatomopathologie , COVID-19/virologie , Protéines d'enveloppe des coronavirus/génétique , Régulation négative/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique , Humains , Inflammasomes/effets des médicaments et des substances chimiques , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Janus kinases/génétique , Janus kinases/métabolisme , Lipopolysaccharides/pharmacologie , Macrophages/cytologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine/déficit , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Poly I-C/pharmacologie , ARN viral/métabolisme , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/isolement et purification
5.
EBioMedicine ; 73: 103672, 2021 Nov.
Article Dans Anglais | MEDLINE | ID: covidwho-1568646

Résumé

BACKGROUND: Phospho-Akt1 (pAkt1) undergoes prolyl hydroxylation at Pro125 and Pro313 by the prolyl hydroxylase-2 (PHD2) in a reaction decarboxylating α-ketoglutarate (αKG). We investigated whether the αKG supplementation could inhibit Akt-mediated activation of platelets and monocytes, in vitro as well as in vivo, by augmenting PHD2 activity. METHODS: We treated platelets or monocytes isolated from healthy individuals with αKG in presence of agonists in vitro and assessed the signalling molecules including pAkt1. We supplemented mice with dietary αKG and estimated the functional responses of platelets and monocytes ex vivo. Further, we investigated the impact of dietary αKG on inflammation and thrombosis in lungs of mice either treated with thrombosis-inducing agent carrageenan or infected with SARS-CoV-2. FINDINGS: Octyl αKG supplementation to platelets promoted PHD2 activity through elevated intracellular αKG to succinate ratio, and reduced aggregation in vitro by suppressing pAkt1(Thr308). Augmented PHD2 activity was confirmed by increased hydroxylated-proline and enhanced binding of PHD2 to pAkt in αKG-treated platelets. Contrastingly, inhibitors of PHD2 significantly increased pAkt1 in platelets. Octyl-αKG followed similar mechanism in monocytes to inhibit cytokine secretion in vitro. Our data also describe a suppressed pAkt1 and reduced activation of platelets and leukocytes ex vivo from mice supplemented with dietary αKG, unaccompanied by alteration in their number. Dietary αKG significantly reduced clot formation and leukocyte accumulation in various organs including lungs of mice treated with thrombosis-inducing agent carrageenan. Importantly, in SARS-CoV-2 infected hamsters, we observed a significant rescue effect of dietary αKG on inflamed lungs with significantly reduced leukocyte accumulation, clot formation and viral load alongside down-modulation of pAkt in the lung of the infected animals. INTERPRETATION: Our study suggests that dietary αKG supplementation prevents Akt-driven maladies such as thrombosis and inflammation and rescues pathology of COVID19-infected lungs. FUNDING: Study was funded by the Department of Biotechnology (DBT), Govt. of India (grants: BT/PR22881 and BT/PR22985); and the Science and Engineering Research Board, Govt. of India (CRG/000092).


Sujets)
Acides cétoglutariques/usage thérapeutique , Prolyl hydroxylases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Thrombose/prévention et contrôle , Animaux , Plaquettes/cytologie , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , COVID-19/anatomopathologie , COVID-19/prévention et contrôle , COVID-19/médecine vétérinaire , COVID-19/virologie , Cricetinae , Compléments alimentaires , Régulation négative/effets des médicaments et des substances chimiques , Humains , Acides cétoglutariques/pharmacologie , Poumon/métabolisme , Poumon/anatomopathologie , Mesocricetus , Souris , Souris de lignée BALB C , Monocytes/cytologie , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Phosphorylation , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Protéines proto-oncogènes c-akt/génétique , SARS-CoV-2/isolement et purification , SARS-CoV-2/physiologie , Thrombose/induit chimiquement , Thrombose/anatomopathologie , Thrombose/médecine vétérinaire
6.
Sci Rep ; 11(1): 23670, 2021 12 08.
Article Dans Anglais | MEDLINE | ID: covidwho-1560986

Résumé

Among cases of SARS-CoV-2 infections that result in serious conditions or death, many have pre-existing conditions such as hypertension and are on renin-angiotensin-aldosterone system (RAAS) inhibitors. The angiotensin-converting-enzyme-2 (ACE2), a key protein of the RAAS pathway, also mediates cellular entry of SARS-CoV-2. RAAS inhibitors might affect the expression levels of ace2, which could impact patient susceptibility to SARS-CoV-2. However, multi-organ-specific information is currently lacking and no species other than rodents have been examined. To address this knowledge gap, we treated adult zebrafish with the RAAS inhibitors aliskiren, olmesartan, and captopril for 7 consecutive days and performed qRT-PCR analysis of major RAAS pathway genes in the brain, gill, heart, intestine, kidney, and liver. Both olmesartan and captopril significantly increased ace2 expression in the heart, gill, and kidney. Olmesartan also increased ace2 expression in the intestine. Conversely, aliskiren significantly decreased ace2 expression in the heart. Discontinuation of compound treatments for 7 days did not return ace2 expression to baseline levels. While potential risks or benefits of antihypertensive RAAS inhibitors to SARS-CoV-2 infections in humans remain uncertain, this study provides new insights regarding the impact of RAAS inhibitors on organ-specific ace2 expression in another vertebrate model, thereby providing comparative data and laying scientific groundwork for future clinical decisions of RAAS inhibitor use in the context of COVID-19.


Sujets)
Angiotensin-converting enzyme 2/métabolisme , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Danio zébré/métabolisme , Amides/pharmacologie , Angiotensin-converting enzyme 2/génétique , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , COVID-19/anatomopathologie , COVID-19/virologie , Fumarates/pharmacologie , Branchies/effets des médicaments et des substances chimiques , Branchies/métabolisme , Humains , Imidazoles/pharmacologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Modèles animaux , SARS-CoV-2/isolement et purification , Tétrazoles/pharmacologie
7.
Int Immunopharmacol ; 101(Pt A): 108264, 2021 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-1487769

Résumé

Topoisomerase (TOP) inhibitors were commonly used as chemotherapeutic agents in the treatment of cancers. In our present study, we found that etoposide (ETO), a topoisomerase 2 (TOP2) inhibitor, upregulated the production of Interleukin 10 (IL-10) in lipopolysaccharide (LPS)-stimulated macrophages. Besides, other TOP2 inhibitors including doxorubicin hydrochloride (DOX) and teniposide (TEN) were also able to augment IL-10 production. Meanwhile, the expression levels of pro-inflammatory factors, for example IL-6 and TNF-α, were also decreased accordingly by the treatment of the TOP2 inhibitors. Of note, ETO facilitated IL-10 secretion, which might be regulated by transcription factor Maf via PI3K/AKT pathway, as pharmaceutic blockage of kinase PI3K or AKT attenuated ETO-induced Maf and IL-10 expression. Further, in LPS-induced mice sepsis model, the enhanced generation of IL-10 was observed in ETO-treated mice, whereas pro-inflammatory cytokines were decreased, which significantly reduced the mortality of mice from LPS-induced lethal cytokine storm. Taken together, these results indicated that ETO may exhibit an anti-inflammatory role by upregulating the alteration of transcription factor Maf and promoting subsequential IL-10 secretion via PI3K/Akt pathway in LPS-induced macrophages. Therefore, ETO may serve as a potential anti-inflammatory agent and employed to severe pro-inflammatory diseases including COVID-19.


Sujets)
Anti-inflammatoires/pharmacologie , Étoposide/pharmacologie , Interleukine-10/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-maf/génétique , Inhibiteurs de la topoisomérase-II/pharmacologie , Animaux , Anti-inflammatoires/usage thérapeutique , Lignée cellulaire , Modèles animaux de maladie humaine , Régulation négative/effets des médicaments et des substances chimiques , Étoposide/usage thérapeutique , Femelle , Interleukine-10/génétique , Interleukine-6/génétique , Interleukine-6/métabolisme , Lipopolysaccharides/toxicité , Macrophages/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Protéines proto-oncogènes c-maf/métabolisme , Choc septique/induit chimiquement , Choc septique/traitement médicamenteux , Inhibiteurs de la topoisomérase-II/usage thérapeutique , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Régulation positive/effets des médicaments et des substances chimiques ,
8.
Cell Res ; 31(12): 1230-1243, 2021 12.
Article Dans Anglais | MEDLINE | ID: covidwho-1475291

Résumé

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the ongoing global pandemic that poses substantial challenges to public health worldwide. A subset of COVID-19 patients experience systemic inflammatory response, known as cytokine storm, which may lead to death. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is an important mediator of inflammation and cell death. Here, we examined the interaction of RIPK1-mediated innate immunity with SARS-CoV-2 infection. We found evidence of RIPK1 activation in human COVID-19 lung pathological samples, and cultured human lung organoids and ACE2 transgenic mice infected by SARS-CoV-2. Inhibition of RIPK1 using multiple small-molecule inhibitors reduced the viral load of SARS-CoV-2 in human lung organoids. Furthermore, therapeutic dosing of the RIPK1 inhibitor Nec-1s reduced mortality and lung viral load, and blocked the CNS manifestation of SARS-CoV-2 in ACE2 transgenic mice. Mechanistically, we found that the RNA-dependent RNA polymerase of SARS-CoV-2, NSP12, a highly conserved central component of coronaviral replication and transcription machinery, promoted the activation of RIPK1. Furthermore, NSP12 323L variant, encoded by the SARS-CoV-2 C14408T variant first detected in Lombardy, Italy, that carries a Pro323Leu amino acid substitution in NSP12, showed increased ability to activate RIPK1. Inhibition of RIPK1 downregulated the transcriptional induction of proinflammatory cytokines and host factors including ACE2 and EGFR that promote viral entry into cells. Our results suggest that SARS-CoV-2 may have an unexpected and unusual ability to hijack the RIPK1-mediated host defense response to promote its own propagation and that inhibition of RIPK1 may provide a therapeutic option for the treatment of COVID-19.


Sujets)
COVID-19/anatomopathologie , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , SARS-CoV-2/physiologie , Angiotensin-converting enzyme 2/génétique , Animaux , COVID-19/mortalité , COVID-19/virologie , ARN polymérase ARN-dépendante de coronavirus/génétique , ARN polymérase ARN-dépendante de coronavirus/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Humains , Imidazoles/pharmacologie , Imidazoles/usage thérapeutique , Indoles/pharmacologie , Indoles/usage thérapeutique , Poumon/anatomopathologie , Poumon/virologie , Souris , Souris transgéniques , Mutation , Receptor-Interacting Protein Serine-Threonine Kinases/antagonistes et inhibiteurs , SARS-CoV-2/isolement et purification , SARS-CoV-2/métabolisme , Taux de survie , Transcriptome/effets des médicaments et des substances chimiques , Charge virale/effets des médicaments et des substances chimiques , Pénétration virale ,
9.
Nat Commun ; 12(1): 5536, 2021 09 20.
Article Dans Anglais | MEDLINE | ID: covidwho-1428813

Résumé

Coronaviruses (CoVs) are important human pathogens for which no specific treatment is available. Here, we provide evidence that pharmacological reprogramming of ER stress pathways can be exploited to suppress CoV replication. The ER stress inducer thapsigargin efficiently inhibits coronavirus (HCoV-229E, MERS-CoV, SARS-CoV-2) replication in different cell types including primary differentiated human bronchial epithelial cells, (partially) reverses the virus-induced translational shut-down, improves viability of infected cells and counteracts the CoV-mediated downregulation of IRE1α and the ER chaperone BiP. Proteome-wide analyses revealed specific pathways, protein networks and components that likely mediate the thapsigargin-induced antiviral state, including essential (HERPUD1) or novel (UBA6 and ZNF622) factors of ER quality control, and ER-associated protein degradation complexes. Additionally, thapsigargin blocks the CoV-induced selective autophagic flux involving p62/SQSTM1. The data show that thapsigargin hits several central mechanisms required for CoV replication, suggesting that this compound (or derivatives thereof) may be developed into broad-spectrum anti-CoV drugs.


Sujets)
Stress du réticulum endoplasmique , SARS-CoV-2/physiologie , Réplication virale/physiologie , Animaux , Autophagie/effets des médicaments et des substances chimiques , Bronches/anatomopathologie , COVID-19/anatomopathologie , COVID-19/virologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Extrait cellulaire , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Chlorocebus aethiops , Coronavirus humain 229E/physiologie , Régulation négative/effets des médicaments et des substances chimiques , Chaperonne BiP du réticulum endoplasmique , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/génétique , Dégradation associée au réticulum endoplasmique/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/virologie , Protéines du choc thermique/métabolisme , Humains , Macrolides/pharmacologie , Coronavirus du syndrome respiratoire du Moyen-Orient/effets des médicaments et des substances chimiques , Coronavirus du syndrome respiratoire du Moyen-Orient/physiologie , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Protéome/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Reproductibilité des résultats , SARS-CoV-2/effets des médicaments et des substances chimiques , Thapsigargine/pharmacologie , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Cellules Vero , Réplication virale/effets des médicaments et des substances chimiques
11.
FASEB J ; 35(9): e21870, 2021 09.
Article Dans Anglais | MEDLINE | ID: covidwho-1373669

Résumé

COVID-19 is often characterized by dysregulated inflammatory and immune responses. It has been shown that the Traditional Chinese Medicine formulation Qing-Fei-Pai-Du decoction (QFPDD) is effective in the treatment of the disease, especially for patients in the early stage. Our network pharmacology analyses indicated that many inflammation and immune-related molecules were the targets of the active components of QFPDD, which propelled us to examine the effects of the decoction on inflammation. We found in the present study that QFPDD effectively alleviated dextran sulfate sodium-induced intestinal inflammation in mice. It inhibited the production of pro-inflammatory cytokines IL-6 and TNFα, and promoted the expression of anti-inflammatory cytokine IL-10 by macrophagic cells. Further investigations found that QFPDD and one of its active components wogonoside markedly reduced LPS-stimulated phosphorylation of transcription factor ATF2, an important regulator of multiple cytokines expression. Our data revealed that both QFPDD and wogonoside decreased the half-life of ATF2 and promoted its proteasomal degradation. Of note, QFPDD and wogonoside down-regulated deubiquitinating enzyme USP14 along with inducing ATF2 degradation. Inhibition of USP14 with the small molecular inhibitor IU1 also led to the decrease of ATF2 in the cells, indicating that QFPDD and wogonoside may act through regulating USP14 to promote ATF2 degradation. To further assess the importance of ubiquitination in regulating ATF2, we generated mice that were intestinal-specific KLHL5 deficiency, a CUL3-interacting protein participating in substrate recognition of E3s. In these mice, QFPDD mitigated inflammatory reaction in the spleen, but not intestinal inflammation, suggesting CUL3-KLHL5 may function as an E3 for ATF2 degradation.


Sujets)
Facteur de transcription ATF-2/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Flavanones/pharmacologie , Glucosides/pharmacologie , Inflammation/traitement médicamenteux , Protéolyse/effets des médicaments et des substances chimiques , Ubiquitin thiolesterase/déficit , Animaux , Lignée cellulaire , Colite/induit chimiquement , Colite/traitement médicamenteux , Cullines/métabolisme , Cytokines/métabolisme , Sulfate dextran/pharmacologie , Sulfate dextran/usage thérapeutique , Médicaments issus de plantes chinoises/usage thérapeutique , Flavanones/usage thérapeutique , Glucosides/usage thérapeutique , Inflammation/induit chimiquement , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Mâle , Souris , Souris de lignée C57BL , Phosphorylation/effets des médicaments et des substances chimiques , Proteasome endopeptidase complex/effets des médicaments et des substances chimiques , Proteasome endopeptidase complex/métabolisme , Pyrroles/pharmacologie , Pyrrolidines/pharmacologie , Ubiquitin thiolesterase/antagonistes et inhibiteurs , Ubiquitination
12.
Adipocyte ; 10(1): 408-411, 2021 12.
Article Dans Anglais | MEDLINE | ID: covidwho-1360282

Résumé

Angiotensin converting enzyme-2 (ACE2) is the cell-surface receptor enabling cellular entry of SARS-CoV-2. ACE2 is highly expressed in adipose tissue (AT), rendering AT a potential SARS-CoV-2 reservoir contributing to massive viral spread in COVID-19 patients with obesity. Although rodent and cell studies suggest that the polyphenol resveratrol alters ACE2, human studies are lacking. Here, we investigated the effects of 30-days resveratrol supplementation on RAS components in AT and skeletal muscle in men with obesity in a placebo-controlled cross-over study. Resveratrol markedly decreased ACE2 (~40%) and leptin (~30%), but did neither alter angiotensinogen, ACE and AT1R expression in AT nor skeletal muscle RAS components. These findings demonstrate that resveratrol supplementation reduces ACE2 in AT, which might dampen SARS-CoV-2 spread in COVID-19.


Sujets)
Tissu adipeux/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Resvératrol/administration et posologie , Tissu adipeux/cytologie , Angiotensin-converting enzyme 2/génétique , COVID-19/anatomopathologie , COVID-19/virologie , Études croisées , Compléments alimentaires , Méthode en double aveugle , Régulation négative/effets des médicaments et des substances chimiques , Humains , Leptine/génétique , Leptine/métabolisme , Mâle , Adulte d'âge moyen , Obésité/traitement médicamenteux , Obésité/anatomopathologie , Effet placebo , Récepteur de type 1 à l'angiotensine-II/génétique , Récepteur de type 1 à l'angiotensine-II/métabolisme , Resvératrol/pharmacologie , SARS-CoV-2/isolement et purification
13.
Eur J Pharmacol ; 908: 174374, 2021 Oct 05.
Article Dans Anglais | MEDLINE | ID: covidwho-1322083

Résumé

The efficacy of corticosteroids and its use for the treatment of SARS-CoV-2 infections is controversial. In this study, using data sets of SARS-CoV-2 infected lung tissues and nasopharyngeal swabs, as well as in vitro experiments, we show that SARS-CoV-2 infection significantly downregulates DUSP1 expression. This downregulation of DUSP1 could be the mechanism regulating the enhanced activation of MAPK pathway as well as the reported steroid resistance in SARS-CoV-2 infection. Moreover, chloroquine, an off labeled COVID-19 drug is able to induce DUSP1 and attenuate MAPK pathway; and is expected to improve sensitivity to steroid treatment. However, further mechanistic studies are required to confirm this effect.


Sujets)
, Chloroquine/pharmacologie , Dual Specificity Phosphatase 1/génétique , Glucocorticoïdes/pharmacologie , p38 Mitogen-Activated Protein Kinases/métabolisme , Adulte , Sujet âgé , COVID-19/anatomopathologie , COVID-19/virologie , Études cas-témoins , Cellules cultivées , Chloroquine/usage thérapeutique , Jeux de données comme sujet , Régulation négative/effets des médicaments et des substances chimiques , Résistance aux substances/effets des médicaments et des substances chimiques , Résistance aux substances/génétique , Synergie des médicaments , Dual Specificity Phosphatase 1/métabolisme , Fibroblastes , Glucocorticoïdes/usage thérapeutique , Volontaires sains , Humains , Poumon/cytologie , Poumon/anatomopathologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/génétique , Adulte d'âge moyen , Partie nasale du pharynx/virologie , Utilisation hors indication , Culture de cellules primaires , SARS-CoV-2/isolement et purification , SARS-CoV-2/pathogénicité
14.
Nat Commun ; 12(1): 4068, 2021 07 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1294463

Résumé

SARS-CoV-2 attacks various organs, most destructively the lung, and cellular entry requires two host cell surface proteins: ACE2 and TMPRSS2. Downregulation of one or both of these is thus a potential therapeutic approach for COVID-19. TMPRSS2 is a known target of the androgen receptor, a ligand-activated transcription factor; androgen receptor activation increases TMPRSS2 levels in various tissues, most notably prostate. We show here that treatment with the antiandrogen enzalutamide-a well-tolerated drug widely used in advanced prostate cancer-reduces TMPRSS2 levels in human lung cells and in mouse lung. Importantly, antiandrogens significantly reduced SARS-CoV-2 entry and infection in lung cells. In support of this experimental data, analysis of existing datasets shows striking co-expression of AR and TMPRSS2, including in specific lung cell types targeted by SARS-CoV-2. Together, the data presented provides strong evidence to support clinical trials to assess the efficacy of antiandrogens as a treatment option for COVID-19.


Sujets)
Antagonistes des androgènes/pharmacologie , Benzamides/pharmacologie , , Nitriles/pharmacologie , 3-Phényl-2-thiohydantoïne/pharmacologie , Serine endopeptidases/métabolisme , Pénétration virale/effets des médicaments et des substances chimiques , Angiotensin-converting enzyme 2/synthèse chimique , Angiotensin-converting enzyme 2/métabolisme , Animaux , COVID-19/métabolisme , COVID-19/virologie , Régulation négative/effets des médicaments et des substances chimiques , Femelle , Humains , Poumon/métabolisme , Poumon/virologie , Mâle , Souris , SARS-CoV-2/effets des médicaments et des substances chimiques , Serine endopeptidases/génétique
15.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article Dans Anglais | MEDLINE | ID: covidwho-1288900

Résumé

A group of clinically approved cancer therapeutic tyrosine kinase inhibitors was screened to test their effects on the expression of angiotensin-converting enzyme 2 (ACE2), the cell surface receptor for SARS-CoV-2. Here, we show that the receptor tyrosine kinase inhibitor imatinib (also known as STI571, Gleevec) can inhibit the expression of the endogenous ACE2 gene at both the transcript and protein levels. Treatment with imatinib resulted in inhibition of cell entry of the viral pseudoparticles (Vpps) in cell culture. In FVB mice orally fed imatinib, tissue expression of ACE2 was reduced, specifically in the lungs and renal tubules, but not in the parenchyma of other organs such as the heart and intestine. Our finding suggests that receptor tyrosine kinases play a role in COVID-19 infection and can be therapeutic targets with combined treatments of the best conventional care of COVID-19.


Sujets)
Angiotensin-converting enzyme 2/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Mésilate d'imatinib/pharmacologie , SARS-CoV-2/physiologie , Pénétration virale/effets des médicaments et des substances chimiques , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/génétique , Animaux , COVID-19/anatomopathologie , COVID-19/virologie , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Chlorocebus aethiops , Femelle , Gènes rapporteurs , Humains , Souris , Régions promotrices (génétique) , SARS-CoV-2/isolement et purification
16.
Life Sci ; 280: 119752, 2021 Sep 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1281493

Résumé

AIMS: Angiotensin-converting enzyme 2 (ACE2) is a key negative regulator of the renin-angiotensin system and also a major receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here, we reveal a role for NF-κB in human lung cell expression of ACE2, and we further explore the potential utility of repurposing NF-κB inhibitors to downregulate ACE2. MAIN METHODS: Expression of ACE2 was assessed by Western blotting and RT-qPCR in multiple human lung cell lines with or without NF-κB inhibitor treatment. Surface ACE2 expression and intracellular reactive oxygen species (ROS) levels were measured with flow cytometry. p50 was knocked down with siRNA. Cytotoxicity was monitored by PARP cleavage and MTS assay. KEY FINDINGS: Pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor, suppressed endogenous ACE2 mRNA and protein expression in H322M and Calu-3 cells. The ROS level in H322M cells was increased after PDTC treatment, and pretreatment with N-acetyl-cysteine (NAC) reversed PDTC-induced ACE2 suppression. Meanwhile, treatment with hydrogen peroxide augmented ACE2 suppression in H322M cells with p50 knockdown. Two repurposed NF-κB inhibitors, the anthelmintic drug triclabendazole and the antiprotozoal drug emetine, also reduced ACE2 mRNA and protein levels. Moreover, zinc supplementation augmented the suppressive effects of triclabendazole and emetine on ACE2 expression in H322M and Calu-3 cells. SIGNIFICANCE: These results suggest that ACE2 expression is modulated by ROS and NF-κB signaling in human lung cells, and the combination of zinc with triclabendazole or emetine shows promise for clinical treatment of ACE2-related disease.


Sujets)
Angiotensin-converting enzyme 2/génétique , Antiparasitaires/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Émétine/pharmacologie , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Triclabendazole/pharmacologie , Zinc/pharmacologie , COVID-19/génétique , Lignée cellulaire , Repositionnement des médicaments , Humains , Poumon/cytologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Pyrrolidines/pharmacologie , Thiocarbamates/pharmacologie ,
17.
Cells ; 10(6)2021 06 07.
Article Dans Anglais | MEDLINE | ID: covidwho-1259431

Résumé

Coronaviruses such as SARS-CoV-2, which is responsible for COVID-19, depend on virus spike protein binding to host cell receptors to cause infection. The SARS-CoV-2 spike protein binds primarily to ACE2 on target cells and is then processed by membrane proteases, including TMPRSS2, leading to viral internalisation or fusion with the plasma membrane. It has been suggested, however, that receptors other than ACE2 may be involved in virus binding. We have investigated the interactions of recombinant versions of the spike protein with human epithelial cell lines that express low/very low levels of ACE2 and TMPRSS2 in a proxy assay for interaction with host cells. A tagged form of the spike protein containing the S1 and S2 regions bound in a temperature-dependent manner to all cell lines, whereas the S1 region alone and the receptor-binding domain (RBD) interacted only weakly. Spike protein associated with cells independently of ACE2 and TMPRSS2, while RBD required the presence of high levels of ACE2 for interaction. As the spike protein has previously been shown to bind heparin, a soluble glycosaminoglycan, we tested the effects of various heparins on ACE2-independent spike protein interaction with cells. Unfractionated heparin inhibited spike protein interaction with an IC50 value of <0.05 U/mL, whereas two low-molecular-weight heparins were less effective. A mutant form of the spike protein, lacking the arginine-rich putative furin cleavage site, interacted only weakly with cells and had a lower affinity for unfractionated and low-molecular-weight heparin than the wild-type spike protein. This suggests that the furin cleavage site might also be a heparin-binding site and potentially important for interactions with host cells. The glycosaminoglycans heparan sulphate and dermatan sulphate, but not chondroitin sulphate, also inhibited the binding of spike protein, indicating that it might bind to one or both of these glycosaminoglycans on the surface of target cells.


Sujets)
Angiotensin-converting enzyme 2/physiologie , Cellules épithéliales/métabolisme , Héparine/pharmacologie , Glycoprotéine de spicule des coronavirus/métabolisme , Cellules A549 , Angiotensin-converting enzyme 2/génétique , Animaux , Sites de fixation/effets des médicaments et des substances chimiques , Sites de fixation/génétique , Cellules Caco-2 , Lignée cellulaire , Chlorocebus aethiops , Chondroïtine sulfate B/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/virologie , Glycosaminoglycanes/pharmacologie , Cellules HEK293 , Cellules HaCaT , Héparitine sulfate/pharmacologie , Humains , Liaison aux protéines/effets des médicaments et des substances chimiques , Liaison aux protéines/génétique , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/physiologie , Glycoprotéine de spicule des coronavirus/composition chimique , Cellules Vero , Pénétration virale/effets des médicaments et des substances chimiques
19.
Viruses ; 13(3)2021 02 27.
Article Dans Anglais | MEDLINE | ID: covidwho-1190473

Résumé

The immunological findings from autopsies, biopsies, and various studies in COVID-19 patients show that the major cause of morbidity and mortality in COVID-19 is excess immune response resulting in hyper-inflammation. With the objective to review various mechanisms of excess immune response in adult COVID-19 patients, Pubmed was searched for free full articles not related to therapeutics or co-morbid sub-groups, published in English until 27.10.2020, irrespective of type of article, country, or region. Joanna Briggs Institute's design-specific checklists were used to assess the risk of bias. Out of 122 records screened for eligibility, 42 articles were included in the final review. The review found that eventually, most mechanisms result in cytokine excess and up-regulation of Nuclear Factor-κB (NF-κB) signaling as a common pathway of excess immune response. Molecules blocking NF-κB or targeting downstream effectors like Tumour Necrosis Factor α (TNFα) are either undergoing clinical trials or lack specificity and cause unwanted side effects. Neutralization of upstream histamine by histamine-conjugated normal human immunoglobulin has been demonstrated to inhibit the nuclear translocation of NF-κB, thereby preventing the release of pro-inflammatory cytokines Interleukin (IL) 1ß, TNF-α, and IL-6 and IL-10 in a safer manner. The authors recommend repositioning it in COVID-19.


Sujets)
COVID-19/immunologie , Syndrome de libération de cytokines/traitement médicamenteux , Syndrome de libération de cytokines/immunologie , Histamine/administration et posologie , Immunoglobulines/administration et posologie , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Facteur de transcription NF-kappa B/immunologie , Syndrome de libération de cytokines/prévention et contrôle , Syndrome de libération de cytokines/virologie , Bases de données factuelles , Régulation négative/effets des médicaments et des substances chimiques , Repositionnement des médicaments , Humains , Immunité , Médicament orphelin , SARS-CoV-2/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
20.
Can J Physiol Pharmacol ; 99(5): 449-460, 2021 May.
Article Dans Anglais | MEDLINE | ID: covidwho-1125455

Résumé

Ribavirin is a guanosine analog with broad-spectrum antiviral activity against RNA viruses. Based on this, we aimed to show the anti-SARS-CoV-2 activity of this drug molecule via in vitro, in silico, and molecular techniques. Ribavirin showed antiviral activity in Vero E6 cells following SARS-CoV-2 infection, whereas the drug itself did not show any toxic effect over the concentration range tested. In silico analysis suggested that ribavirin has a broad-spectrum impact on SARS-CoV-2, acting at different viral proteins. According to the detailed molecular techniques, ribavirin was shown to decrease the expression of TMPRSS2 at both mRNA and protein levels 48 h after treatment. The suppressive effect of ribavirin in ACE2 protein expression was shown to be dependent on cell types. Finally, proteolytic activity assays showed that ribavirin also showed an inhibitory effect on the TMPRSS2 enzyme. Based on these results, we hypothesized that ribavirin may inhibit the expression of TMPRSS2 by modulating the formation of inhibitory G-quadruplex structures at the TMPRSS2 promoter. As a conclusion, ribavirin is a potential antiviral drug for the treatment against SARS-CoV-2, and it interferes with the effects of TMPRSS2 and ACE2 expression.


Sujets)
Angiotensin-converting enzyme 2/métabolisme , Antiviraux/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Ribavirine/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Serine endopeptidases/métabolisme , Animaux , Cellules Caco-2 , Chlorocebus aethiops , G-quadruplexes/effets des médicaments et des substances chimiques , Humains , Régions promotrices (génétique)/génétique , SARS-CoV-2/physiologie , Serine endopeptidases/génétique , Cellules Vero
SÉLECTION CITATIONS
Détails de la recherche