Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.684
Filtrer
1.
Nat Commun ; 15(1): 5503, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951531

RÉSUMÉ

Proline is widely known as the only proteogenic amino acid with a secondary amine. In addition to its crucial role in protein structure, the secondary amino acid modulates neurotransmission and regulates the kinetics of signaling proteins. To understand the structural basis of proline import, we solved the structure of the proline transporter SIT1 in complex with the COVID-19 viral receptor ACE2 by cryo-electron microscopy. The structure of pipecolate-bound SIT1 reveals the specific sequence requirements for proline transport in the SLC6 family and how this protein excludes amino acids with extended side chains. By comparing apo and substrate-bound SIT1 states, we also identify the structural changes that link substrate release and opening of the cytoplasmic gate and provide an explanation for how a missense mutation in the transporter causes iminoglycinuria.


Sujet(s)
Angiotensin-converting enzyme 2 , Cryomicroscopie électronique , Proline , SARS-CoV-2 , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/génétique , Proline/métabolisme , Humains , SARS-CoV-2/métabolisme , SARS-CoV-2/génétique , COVID-19/virologie , COVID-19/métabolisme , Systèmes de transport d'acides aminés neutres/métabolisme , Systèmes de transport d'acides aminés neutres/génétique , Systèmes de transport d'acides aminés neutres/composition chimique , Modèles moléculaires
2.
Ethiop J Health Sci ; 34(1): 85-100, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38957334

RÉSUMÉ

Background: Coronaviruses (CoVs) belong to the RNA viruses family. The viruses in this family are known to cause mild respiratory disease in humans. The origin of the novel SARS-COV2 virus that caused the coronavirus-19 disease (COVID-19) is the Wuhan city in China from where it disseminated to cause a global pandemic. Although lungs are the predominant target organ for Coronavirus Disease-19 (COVID-19), since its outbreak, the disease is known to affect heart, blood vessels, kidney, intestine, liver and brain. This review aimed to summarize the catastrophic impacts of Coronavirus disease-19 on heart and liver along with its mechanisms of pathogenesis. Methods: The information used in this review was obtained from relevant articles published on PubMed, Google Scholar, Google, WHO website, CDC and other sources. Key searching statements and phrases related to COVID-19 were used to retrieve information. Original research articles, review papers, research letters and case reports were used as a source of information. Results: Besides causing severe lung injury, COVID-19 has also been reported to affect and cause dysfunction of many other organs. COVID-19 infection can affect people by downregulating membrane-bound active angiotensin-converting enzyme (ACE). People who have deficient ACE2 expression are more vulnerable to COVID-19 infection. The patients' pre-existing co-morbidities are major risk factors that predispose individuals to severe COVID-19. Conclusion: The disease severity and its broad spectrum phenotype is a result of combined direct and indirect pathogenic factors. Therefore, protocols that harmonize many therapeutic preferences should be the best alternatives to de-escalate the disease and obviate deaths caused as a result of multiple organ damage and dysfunction induced by the disease.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , COVID-19/complications , COVID-19/épidémiologie , Maladies du foie/étiologie , Maladies du foie/virologie , Cardiopathies/étiologie , Cardiopathies/virologie , Angiotensin-converting enzyme 2/métabolisme , Foie/anatomopathologie , Foie/virologie
3.
J Gen Virol ; 105(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38995681

RÉSUMÉ

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is associated with neurological sequelae including haemorrhage, thrombosis and ischaemic necrosis and encephalitis. However, the mechanism by which this occurs is unclear. Neurological disease associated with COVID-19 has been proposed to occur following direct infection of the central nervous system and/or indirectly by local or systemic immune activation. We evaluated the expression of angiotensin-converting enzyme-2 and transmembrane protease, serine 2 (TMPRSS2) in brain tissue from five healthy human donors and observed low-level expression of these proteins in cells morphologically consistent with astrocytes, neurons and choroidal ependymal cells within the frontal cortex and medulla oblongata. Primary human astrocytes, neurons, choroid plexus epithelial cells and pericytes supported productive SARS-CoV-2 infection with ancestral, Alpha, Delta and Omicron variants. Infected cells supported the full viral life cycle, releasing infectious virus particles. In contrast, primary brain microvascular endothelial cells and microglia were refractory to SARS-CoV-2 infection. These data support a model whereby SARS-CoV-2 can infect human brain cells, and the mechanism of viral entry warrants further investigation.


Sujet(s)
Angiotensin-converting enzyme 2 , Astrocytes , COVID-19 , Plexus choroïde , Cellules épithéliales , Neurones , Péricytes , SARS-CoV-2 , Serine endopeptidases , Humains , Péricytes/virologie , SARS-CoV-2/physiologie , Astrocytes/virologie , Plexus choroïde/virologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Neurones/virologie , COVID-19/virologie , COVID-19/anatomopathologie , Cellules épithéliales/virologie , Serine endopeptidases/métabolisme , Serine endopeptidases/génétique , Cellules cultivées , Encéphale/virologie , Encéphale/anatomopathologie , Système nerveux central/virologie
4.
Sci Immunol ; 9(97): eadn0178, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996010

RÉSUMÉ

Virus-induced cell death is a key contributor to COVID-19 pathology. Cell death induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is well studied in myeloid cells but less in its primary host cell type, angiotensin-converting enzyme 2 (ACE2)-expressing human airway epithelia (HAE). SARS-CoV-2 induces apoptosis, necroptosis, and pyroptosis in HAE organotypic cultures. Single-cell and limiting-dilution analysis revealed that necroptosis is the primary cell death event in infected cells, whereas uninfected bystanders undergo apoptosis, and pyroptosis occurs later during infection. Mechanistically, necroptosis is induced by viral Z-RNA binding to Z-DNA-binding protein 1 (ZBP1) in HAE and lung tissues from patients with COVID-19. The Delta (B.1.617.2) variant, which causes more severe disease than Omicron (B1.1.529) in humans, is associated with orders of magnitude-greater Z-RNA/ZBP1 interactions, necroptosis, and disease severity in animal models. Thus, Delta induces robust ZBP1-mediated necroptosis and more disease severity.


Sujet(s)
COVID-19 , Nécroptose , Pyroptose , Protéines de liaison à l'ARN , Muqueuse respiratoire , SARS-CoV-2 , Humains , SARS-CoV-2/immunologie , COVID-19/immunologie , COVID-19/anatomopathologie , Nécroptose/immunologie , Animaux , Muqueuse respiratoire/virologie , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/anatomopathologie , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris , Mort cellulaire/immunologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Apoptose/immunologie
5.
Pharmacol Res Perspect ; 12(4): e1244, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38982716

RÉSUMÉ

We hypothesize that a "Faustian bargain"-the trading of increased SARS-CoV2 viral infection with a concurrent potential for prevention of life-threatening lower lung infection explains the previous and future morbidity and mortality from COVID-19. Further, this trade-off is made feasible by fundamental principles of thermodynamics and receptor affinity.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , COVID-19/virologie , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Thermodynamique , Angiotensin-converting enzyme 2/métabolisme
6.
Front Cell Infect Microbiol ; 14: 1371837, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994005

RÉSUMÉ

Virus receptors determine the tissue tropism of viruses and have a certain relationship with the clinical outcomes caused by viral infection, which is of great importance for the identification of virus receptors to understand the infection mechanism of viruses and to develop entry inhibitor. Proximity labeling (PL) is a new technique for studying protein-protein interactions, but it has not yet been applied to the identification of virus receptors or co-receptors. Here, we attempt to identify co-receptor of SARS-CoV-2 by employing TurboID-catalyzed PL. The membrane protein angiotensin-converting enzyme 2 (ACE2) was employed as a bait and conjugated to TurboID, and a A549 cell line with stable expression of ACE2-TurboID was constructed. SARS-CoV-2 pseudovirus were incubated with ACE2-TurboID stably expressed cell lines in the presence of biotin and ATP, which could initiate the catalytic activity of TurboID and tag adjacent endogenous proteins with biotin. Subsequently, the biotinylated proteins were harvested and identified by mass spectrometry. We identified a membrane protein, AXL, that has been functionally shown to mediate SARS-CoV-2 entry into host cells. Our data suggest that PL could be used to identify co-receptors for virus entry.


Sujet(s)
Angiotensin-converting enzyme 2 , Récepteurs viraux , SARS-CoV-2 , Pénétration virale , Humains , Angiotensin-converting enzyme 2/métabolisme , SARS-CoV-2/métabolisme , SARS-CoV-2/physiologie , Cellules A549 , Récepteurs viraux/métabolisme , Axl Receptor Tyrosine Kinase , Récepteurs à activité tyrosine kinase/métabolisme , Protéines proto-oncogènes/métabolisme , COVID-19/virologie , COVID-19/métabolisme , Coloration et marquage/méthodes , Cellules HEK293 , Biotinylation , Cartographie d'interactions entre protéines , Biotine/métabolisme
7.
Molecules ; 29(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38998974

RÉSUMÉ

Considering the high evolutionary rate and great harmfulness of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is imperative to develop new pharmacological antagonists. Human angiotensin-converting enzyme-2 (ACE2) functions as a primary receptor for the spike protein (S protein) of SARS-CoV-2. Thus, a novel functional peptide, KYPAY (K5), with a boomerang structure, was developed to inhibit the interaction between ACE2 and the S protein by attaching to the ACE2 ligand-binding domain (LBD). The inhibition property of K5 was evaluated via molecular simulations, cell experiments, and adsorption kinetics analysis. The molecular simulations showed that K5 had a high affinity for ACE2 but a low affinity for the cell membrane. The umbrella sampling (US) simulations revealed a significant enhancement in the binding potential of this functional peptide to ACE2. The fluorescence microscopy and cytotoxicity experiments showed that K5 effectively prevented the interaction between ACE2 and the S protein without causing any noticeable harm to cells. Further flow cytometry research indicated that K5 successfully hindered the interaction between ACE2 and the S protein, resulting in 78% inhibition at a concentration of 100 µM. This work offers an innovative perspective on the development of functional peptides for the prevention and therapy of SARS-CoV-2.


Sujet(s)
Angiotensin-converting enzyme 2 , Peptides , Liaison aux protéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , Humains , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/composition chimique , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/métabolisme , Peptides/composition chimique , Peptides/pharmacologie , Simulation de dynamique moléculaire , COVID-19/virologie , COVID-19/métabolisme , Traitements médicamenteux de la COVID-19 , Antiviraux/composition chimique , Antiviraux/pharmacologie , Cinétique
8.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38999930

RÉSUMÉ

Although SARS-CoV-2 induces mucin hypersecretion in the respiratory tract, hyposalivation/xerostomia has been reported by COVID-19 patients. We evaluate the submandibular gland (SMGs) pathogenesis in SARS-CoV-2-infected K18-hACE2 mice, focusing on the impact of infection on the mucin production and structural integrity of acini, ductal system, myoepithelial cells (MECs) and telocytes. The spike protein, the nucleocapsid protein, hACE2, actin, EGF, TNF-α and IL-1ß were detected by immunofluorescence, and the Egfr and Muc5b expression was evaluated. In the infected animals, significant acinar hypertrophy was observed in contrast to ductal atrophy. Nucleocapsid proteins and/or viral particles were detected in the SMG cells, mainly in the nuclear membrane-derived vesicles, confirming the nuclear role in the viral formation. The acinar cells showed intense TNF-α and IL-1ß immunoexpression, and the EGF-EGFR signaling increased, together with Muc5b upregulation. This finding explains mucin hypersecretion and acinar hypertrophy, which compress the ducts. Dying MECs and actin reduction were also observed, indicating failure of contraction and acinar support, favoring acinar hypertrophy. Viral assembly was found in the dying telocytes, pointing to these intercommunicating cells as viral transmitters in SMGs. Therefore, EGF-EGFR-induced mucin hypersecretion was triggered by SARS-CoV-2 in acinar cells, likely mediated by cytokines. The damage to telocytes and MECs may have favored the acinar hypertrophy, leading to ductal obstruction, explaining xerostomia in COVID-19 patients. Thus, acinar cells, telocytes and MECs may be viral targets, which favor replication and cell-to-cell viral transmission in the SMG, corroborating the high viral load in saliva of infected individuals.


Sujet(s)
COVID-19 , Récepteurs ErbB , SARS-CoV-2 , Glande submandibulaire , Xérostomie , COVID-19/anatomopathologie , COVID-19/virologie , COVID-19/métabolisme , Animaux , Glande submandibulaire/virologie , Glande submandibulaire/anatomopathologie , Glande submandibulaire/métabolisme , SARS-CoV-2/physiologie , Souris , Xérostomie/étiologie , Xérostomie/anatomopathologie , Xérostomie/virologie , Xérostomie/métabolisme , Récepteurs ErbB/métabolisme , Humains , Angiotensin-converting enzyme 2/métabolisme , Mucine 5B/métabolisme , Cellules acineuses/anatomopathologie , Cellules acineuses/métabolisme , Cellules acineuses/virologie , Interleukine-1 bêta/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Modèles animaux de maladie humaine
9.
Sci Rep ; 14(1): 15351, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38961189

RÉSUMÉ

As SARS-CoV-2 continues to spread worldwide, tractable primary airway cell models that recapitulate the cell-intrinsic response to arising viral variants are needed. Here we describe an adult stem cell-derived human airway organoid model overexpressing the ACE2 receptor (ACE2-OE) that supports robust viral replication while maintaining 3D architecture and cellular diversity of the airway epithelium. ACE2-OE organoids were infected with SARS-CoV-2 variants and subjected to single-cell RNA-sequencing. Interferon-lambda was upregulated in cells with low-level infection while the NF-kB inhibitor alpha gene (encoding IkBa) was consistently upregulated in infected cells, and its expression positively correlated with infection levels. Confocal microscopy showed more IkBa expression in infected than bystander cells, but found concurrent nuclear translocation of NF-kB that IkBa usually prevents. Overexpressing a nondegradable IkBa mutant reduced NF-kB translocation and increased viral infection. These data demonstrate the functionality of ACE2-OE organoids in SARS-CoV-2 research and underscore that the strength of the NF-kB feedback loop in infected cells controls viral replication.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Inhibiteur alpha de NF-KappaB , Organoïdes , SARS-CoV-2 , Réplication virale , Humains , Organoïdes/virologie , Organoïdes/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , SARS-CoV-2/physiologie , COVID-19/virologie , COVID-19/métabolisme , COVID-19/génétique , Inhibiteur alpha de NF-KappaB/métabolisme , Inhibiteur alpha de NF-KappaB/génétique , Facteur de transcription NF-kappa B/métabolisme
10.
Proc Natl Acad Sci U S A ; 121(30): e2321972121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39008677

RÉSUMÉ

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection inhibits mitochondrial oxidative phosphorylation (OXPHOS) and elevates mitochondrial reactive oxygen species (ROS, mROS) which activates hypoxia-inducible factor-1alpha (HIF-1α), shifting metabolism toward glycolysis to drive viral biogenesis but also causing the release of mitochondrial DNA (mtDNA) and activation of innate immunity. To determine whether mitochondrially targeted antioxidants could mitigate these viral effects, we challenged mice expressing human angiotensin-converting enzyme 2 (ACE2) with SARS-CoV-2 and intervened using transgenic and pharmacological mitochondrially targeted catalytic antioxidants. Transgenic expression of mitochondrially targeted catalase (mCAT) or systemic treatment with EUK8 decreased weight loss, clinical severity, and circulating levels of mtDNA; as well as reduced lung levels of HIF-1α, viral proteins, and inflammatory cytokines. RNA-sequencing of infected lungs revealed that mCAT and Eukarion 8 (EUK8) up-regulated OXPHOS gene expression and down-regulated HIF-1α and its target genes as well as innate immune gene expression. These data demonstrate that SARS-CoV-2 pathology can be mitigated by catalytically reducing mROS, potentially providing a unique host-directed pharmacological therapy for COVID-19 which is not subject to viral mutational resistance.


Sujet(s)
Antioxydants , COVID-19 , Souris transgéniques , Mitochondries , Phosphorylation oxydative , SARS-CoV-2 , Animaux , Souris , COVID-19/virologie , COVID-19/métabolisme , COVID-19/immunologie , COVID-19/anatomopathologie , Antioxydants/métabolisme , Antioxydants/pharmacologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , SARS-CoV-2/effets des médicaments et des substances chimiques , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Humains , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Poumon/virologie , Poumon/anatomopathologie , Poumon/métabolisme , Espèces réactives de l'oxygène/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , ADN mitochondrial/génétique , ADN mitochondrial/métabolisme , Catalase/métabolisme , Catalase/génétique , Traitements médicamenteux de la COVID-19 , Modèles animaux de maladie humaine , Immunité innée
11.
J Med Virol ; 96(7): e29782, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39011762

RÉSUMÉ

Extracellular vesicles (EVs) are shown to be a novel viral transmission model capable of increasing a virus's tropism. According to our earlier research, cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or transfected with envelope protein plasmids generate a novel type of EVs that are micrometer-sized and able to encase virus particles. Here, we showed the capacity of these EVs to invade various animals both in vitro and in vivo independent of the angiotensin-converting enzyme 2 receptor. First, via macropinocytosis, intact EVs produced from Vero E6 (monkey) cells were able to enter cells from a variety of animals, including cats, dogs, bats, hamsters, and minks, and vice versa. Second, when given to zebrafish with cutaneous wounds, the EVs showed favorable stability in aqueous environments and entered the fish. Moreover, infection of wild-type (WT) mice with heterogeneous EVs carrying SARS-CoV-2 particles led to a strong cytokine response and a notable amount of lung damage. Conversely, free viral particles did not infect WT mice. These results highlight the variety of processes behind viral transmission and cross-species evolution by indicating that EVs may be possible vehicles for SARS-CoV-2 spillover and raising risk concerns over EVs' potential for viral gene transfer.


Sujet(s)
COVID-19 , Vésicules extracellulaires , SARS-CoV-2 , Animaux , Vésicules extracellulaires/virologie , Vésicules extracellulaires/métabolisme , SARS-CoV-2/physiologie , SARS-CoV-2/pathogénicité , SARS-CoV-2/génétique , COVID-19/transmission , COVID-19/virologie , Souris , Chlorocebus aethiops , Cellules Vero , Humains , Cricetinae , Protéines d'enveloppe des coronavirus/métabolisme , Protéines d'enveloppe des coronavirus/génétique , Chiens , Danio zébré/virologie , Chats , Chiroptera/virologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique
12.
Protein Sci ; 33(8): e5109, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38989563

RÉSUMÉ

Understanding how proteins evolve under selective pressure is a longstanding challenge. The immensity of the search space has limited efforts to systematically evaluate the impact of multiple simultaneous mutations, so mutations have typically been assessed individually. However, epistasis, or the way in which mutations interact, prevents accurate prediction of combinatorial mutations based on measurements of individual mutations. Here, we use artificial intelligence to define the entire functional sequence landscape of a protein binding site in silico, and we call this approach Complete Combinatorial Mutational Enumeration (CCME). By leveraging CCME, we are able to construct a comprehensive map of the evolutionary connectivity within this functional sequence landscape. As a proof of concept, we applied CCME to the ACE2 binding site of the SARS-CoV-2 spike protein receptor binding domain. We selected representative variants from across the functional sequence landscape for testing in the laboratory. We identified variants that retained functionality to bind ACE2 despite changing over 40% of evaluated residue positions, and the variants now escape binding and neutralization by monoclonal antibodies. This work represents a crucial initial stride toward achieving precise predictions of pathogen evolution, opening avenues for proactive mitigation.


Sujet(s)
Angiotensin-converting enzyme 2 , Mutation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/génétique , SARS-CoV-2/génétique , SARS-CoV-2/composition chimique , SARS-CoV-2/métabolisme , Humains , Sites de fixation , COVID-19/virologie , COVID-19/génétique , Liaison aux protéines , Intelligence artificielle
13.
Int J Nanomedicine ; 19: 6931-6943, 2024.
Article de Anglais | MEDLINE | ID: mdl-39005960

RÉSUMÉ

Purpose: Over the past three years, extensive research has been dedicated to understanding and combating COVID-19. Targeting the interaction between the SARS-CoV-2 Spike protein and the ACE2 receptor has emerged as a promising therapeutic strategy against SARS-CoV-2. This study aimed to develop ACE2-coated virus-like particles (ACE2-VLPs), which can be utilized to prevent viral entry into host cells and efficiently neutralize the virus. Methods: Virus-like particles were generated through the utilization of a packaging plasmid in conjunction with a plasmid containing the ACE2 envelope sequence. Subsequently, ACE2-VLPs and ACE2-EVs were purified via ultracentrifugation. The quantification of VLPs was validated through multiple methods, including Nanosight 3000, TEM imaging, and Western blot analysis. Various packaging systems were explored to optimize the ACE2-VLP configuration for enhanced neutralization capabilities. The evaluation of neutralization effectiveness was conducted using pseudoviruses bearing different spike protein variants. Furthermore, the study assessed the neutralization potential against the Omicron BA.1 variant in Vero E6 cells. Results: ACE2-VLPs showed a high neutralization capacity even at low doses and demonstrated superior efficacy in in vitro pseudoviral assays compared to extracellular vesicles carrying ACE2. ACE2-VLPs remained stable under various environmental temperatures and effectively blocked all tested variants of concern in vitro. Notably, they exhibited significant neutralization against Omicron BA.1 variant in Vero E6 cells. Given their superior efficacy compared to extracellular vesicles and proven success against live virus, ACE2-VLPs stand out as crucial candidates for treating SARS-CoV-2 infections. Conclusion: This novel therapeutic approach of coating VLPs with receptor particles provides a proof-of-concept for designing effective neutralization strategies for other viral diseases in the future.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Angiotensin-converting enzyme 2/métabolisme , Animaux , Cellules Vero , Chlorocebus aethiops , Humains , COVID-19/virologie , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Anticorps neutralisants/pharmacologie , Cellules HEK293 , Pénétration virale/effets des médicaments et des substances chimiques
14.
Cell Commun Signal ; 22(1): 349, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965547

RÉSUMÉ

T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.


Sujet(s)
Antigènes CD147 , COVID-19 , Lymphopénie , SARS-CoV-2 , Humains , Lymphopénie/immunologie , Lymphopénie/virologie , COVID-19/immunologie , COVID-19/virologie , COVID-19/anatomopathologie , SARS-CoV-2/métabolisme , Antigènes CD147/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/virologie , Syndrome de libération de cytokines/immunologie , Animaux
15.
BMC Infect Dis ; 24(1): 663, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38956476

RÉSUMÉ

BACKGROUND: Severe COVID-19 is uncommon, restricted to 19% of the total population. In response to the first virus wave (alpha variant of SARS-CoV-2), we investigated whether a biomarker indicated severity of disease and, in particular, if variable expression of angiotensin converting enzyme 2 (ACE2) in blood might clarify this difference in risk and of post COVID -19 conditions (PCC). METHODS: The IRB-approved study compared patients hospitalized with severe COVID-19 to healthy controls. Severe infection was defined requiring oxygen or increased oxygen need from baseline at admission with positive COVID-19 PCR. A single blood sample was obtained from patients within a day of admission. ACE2 RNA expression in blood cells was measured by an RT-PCR assay. Plasma ACE1 and ACE2 enzyme activities were quantified by fluorescent peptides. Plasma TIMP-1, PIIINP and MMP-9 antigens were quantified by ELISA. Data were entered into REDCap and analyzed using STATA v 14 and GraphPad Prism v 10. RESULTS: Forty-eight patients and 72 healthy controls were recruited during the pandemic. ACE2 RNA expression in peripheral blood mononuclear cells (PBMC) was rarely detected acutely during severe COVID-19 but common in controls (OR for undetected ACE2: 12.4 [95% CI: 2.62-76.1]). ACE2 RNA expression in PBMC did not determine plasma ACE1 and ACE2 activity, suggesting alternative cell-signaling pathways. Markers of fibrosis (TIMP-1 and PIIINP) and vasculopathy (MMP-9) were additionally elevated. ACE2 RNA expression during severe COVID-19 often responded within hours to convalescent plasma. Analogous to oncogenesis, we speculate that potent, persistent, cryptic processes following COVID-19 (the renin-angiotensin system (RAS), fibrosis and vasculopathy) initiate or promote post-COVID-19 conditions (PCC) in susceptible individuals. CONCLUSIONS: This work elucidates biological and temporal plausibility for ACE2, TIMP1, PIIINP and MMP-9 in the pathogenesis of PCC. Intersection of these independent systems is uncommon and may in part explain the rarity of PCC.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Agranulocytes , SARS-CoV-2 , Humains , COVID-19/sang , Angiotensin-converting enzyme 2/sang , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Agranulocytes/métabolisme , Agranulocytes/virologie , Sujet âgé , Adulte , Marqueurs biologiques/sang , Inhibiteur tissulaire de métalloprotéinase-1/sang , Inhibiteur tissulaire de métalloprotéinase-1/génétique , Matrix metalloproteinase 9/sang , Matrix metalloproteinase 9/génétique , Indice de gravité de la maladie , Études cas-témoins , Peptidyl-Dipeptidase A/sang , Peptidyl-Dipeptidase A/génétique
16.
Front Cell Infect Microbiol ; 14: 1394721, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975331

RÉSUMÉ

Since 2019, Coronavirus Disease 2019(COVID-19) has affected millions of people worldwide. Except for acute respiratory distress syndrome, dysgeusis is also a common symptom of COVID-19 that burdens patients for weeks or permanently. However, the mechanisms underlying taste dysfunctions remain unclear. Here, we performed complete autopsies of five patients who died of COVID-19. Integrated tongue samples, including numerous taste buds, salivary glands, vessels, and nerves were collected to map the pathology, distribution, cell tropism, and receptor distribution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the tongue. Our results revealed that all patients had moderate lymphocyte infiltration around the salivary glands and in the lamina propria adjacent to the mucosa, and pyknosis in the epithelia of taste buds and salivary glands. This may be because the serous acini, salivary gland ducts, and taste buds are the primary sites of SARS-CoV-2 infection. Multicolor immunofluorescence showed that SARS-CoV-2 readily infects Keratin (KRT)7+ taste receptor cells in taste buds, secretory cells in serous acini, and inner epithelial cells in the ducts. The major receptors, angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine subtype 2 (TMPRSS2), were both abundantly expressed in these cells. Viral antigens and receptor were both rarely detected in vessels and nerves. This indicates that SARS-CoV-2 infection triggers pathological injury in the tongue, and that dysgeusis may be directly related to viral infection and cellular damage.


Sujet(s)
Angiotensin-converting enzyme 2 , Autopsie , COVID-19 , SARS-CoV-2 , Serine endopeptidases , Langue , Tropisme viral , Humains , COVID-19/anatomopathologie , COVID-19/virologie , SARS-CoV-2/pathogénicité , Langue/virologie , Langue/anatomopathologie , Mâle , Angiotensin-converting enzyme 2/métabolisme , Femelle , Adulte d'âge moyen , Serine endopeptidases/métabolisme , Glandes salivaires/virologie , Glandes salivaires/anatomopathologie , Sujet âgé , Calicules gustatifs/virologie , Calicules gustatifs/anatomopathologie , Récepteurs viraux/métabolisme
17.
Anal Chim Acta ; 1317: 342919, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39030015

RÉSUMÉ

The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in widespread disease transmission, challenging the stability of global healthcare systems. Surface-enhanced Raman scattering (SERS) as an easy operation, fast, and low-cost technology illustrates a good potential in detecting SARS-CoV-2. In the study, one-step fabrication of gold-silver alloy nanoparticles (AuAgNPs) with adjustable metal proportions and diameters is employed as SERS substrates. The angiotensin-converting enzyme 2 (ACE2) functionalized AuAgNPs are applied as sensor surfaces to detect SARS-CoV-2 S protein. By optimizing the SERS substrates, ACE2/Au35Ag65NPs illustrate higher performance in detecting the SARS-CoV-2 S protein with a limit of detection (LOD) of 10 fg/mL in both phosphate-buffered saline (PBS) and pharyngeal swabs solution (PSS). It also provides excellent reproducibility with a relative standard deviation (RSD) of 7.7 % and 7.9 %, respectively. This easily preparable and highly reproducible SERS substrate has good potential in the practical application of detecting SARS-CoV-2.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Or , Limite de détection , Nanoparticules métalliques , SARS-CoV-2 , Argent , Analyse spectrale Raman , Glycoprotéine de spicule des coronavirus , Analyse spectrale Raman/méthodes , Argent/composition chimique , Glycoprotéine de spicule des coronavirus/analyse , Nanoparticules métalliques/composition chimique , SARS-CoV-2/isolement et purification , Humains , Or/composition chimique , COVID-19/diagnostic , COVID-19/virologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/composition chimique , Alliages/composition chimique
18.
Physiol Rep ; 12(14): e16161, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39020498

RÉSUMÉ

While under physiological conditions angiotensin-converting enzyme 2 (ACE2) is an antagonist of vasoconstrictive agents in the renin-angiotensin-aldosterone system (RAAS), in the context of SARS coronavirus 2 (SARS-CoV-2) ACE2 serves as the gateway into cells. Furthermore, RAAS has previously been shown to be influenced by exercise training and is suggested to be involved in skeletal muscle mass maintenance. Given this connection, the investigation of circulating ACE2 plasma protein concentration before and following acute and chronic endurance and resistance exercise could increase the understanding of the implications of the exposure of athletes to SARS-CoV-2. Therefore, this study investigated levels of circulating ACE2 in lifelong high-level trained endurance and resistance athletes and control subjects in response to either acute endurance or resistance exercise. Results show no baseline differences in absolute ACE2 concentration between groups, but a strong negative correlation with levels of fitness and positive correlation with BMI in control subjects. Furthermore, acute endurance exercise significantly increased ACE2 levels across all groups, but only in the strength group in response to resistance exercise. This indicates that circulating ACE2 plasma levels are influenced by levels of fitness and health, and that acute endurance exercise has a stronger effect on plasma ACE2 levels than resistance exercise.


Sujet(s)
Angiotensin-converting enzyme 2 , Athlètes , Aptitude physique , Humains , Mâle , Angiotensin-converting enzyme 2/sang , Angiotensin-converting enzyme 2/métabolisme , Adulte , Aptitude physique/physiologie , Exercice physique/physiologie , Marqueurs biologiques/sang , COVID-19/sang , Entraînement en résistance/méthodes , Endurance physique/physiologie , Jeune adulte
19.
Proc Natl Acad Sci U S A ; 121(30): e2408109121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39028694

RÉSUMÉ

The prevalence of "long COVID" is just one of the conundrums highlighting how little we know about the lung's response to viral infection, particularly to syndromecoronavirus-2 (SARS-CoV-2), for which the lung is the point of entry. We used an in vitro human lung system to enable a prospective, unbiased, sequential single-cell level analysis of pulmonary cell responses to infection by multiple SARS-CoV-2 strains. Starting with human induced pluripotent stem cells and emulating lung organogenesis, we generated and infected three-dimensional, multi-cell-type-containing lung organoids (LOs) and gained several unexpected insights. First, SARS-CoV-2 tropism is much broader than previously believed: Many lung cell types are infectable, if not through a canonical receptor-mediated route (e.g., via Angiotensin-converting encyme 2(ACE2)) then via a noncanonical "backdoor" route (via macropinocytosis, a form of endocytosis). Food and Drug Administration (FDA)-approved endocytosis blockers can abrogate such entry, suggesting adjunctive therapies. Regardless of the route of entry, the virus triggers a lung-autonomous, pulmonary epithelial cell-intrinsic, innate immune response involving interferons and cytokine/chemokine production in the absence of hematopoietic derivatives. The virus can spread rapidly throughout human LOs resulting in mitochondrial apoptosis mediated by the prosurvival protein Bcl-xL. This host cytopathic response to the virus may help explain persistent inflammatory signatures in a dysfunctional pulmonary environment of long COVID. The host response to the virus is, in significant part, dependent on pulmonary Surfactant Protein-B, which plays an unanticipated role in signal transduction, viral resistance, dampening of systemic inflammatory cytokine production, and minimizing apoptosis. Exogenous surfactant, in fact, can be broadly therapeutic.


Sujet(s)
COVID-19 , Poumon , Organoïdes , SARS-CoV-2 , Pénétration virale , Humains , SARS-CoV-2/physiologie , SARS-CoV-2/immunologie , COVID-19/immunologie , COVID-19/virologie , Poumon/virologie , Poumon/immunologie , Poumon/anatomopathologie , Organoïdes/virologie , Traitements médicamenteux de la COVID-19 , Cellules souches pluripotentes induites/virologie , Angiotensin-converting enzyme 2/métabolisme , Inflammation , Cytokines/métabolisme , Apoptose
20.
Proc Natl Acad Sci U S A ; 121(29): e2310421121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38976733

RÉSUMÉ

We generated a replication-competent OC43 human seasonal coronavirus (CoV) expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in place of the native spike (rOC43-CoV2 S). This virus is highly attenuated relative to OC43 and SARS-CoV-2 in cultured cells and animals and is classified as a biosafety level 2 (BSL-2) agent by the NIH biosafety committee. Neutralization of rOC43-CoV2 S and SARS-CoV-2 by S-specific monoclonal antibodies and human sera is highly correlated, unlike recombinant vesicular stomatitis virus-CoV2 S. Single-dose immunization with rOC43-CoV2 S generates high levels of neutralizing antibodies against SARS-CoV-2 and fully protects human ACE2 transgenic mice from SARS-CoV-2 lethal challenge, despite nondetectable replication in respiratory and nonrespiratory organs. rOC43-CoV2 S induces S-specific serum and airway mucosal immunoglobulin A and IgG responses in rhesus macaques. rOC43-CoV2 S has enormous value as a BSL-2 agent to measure S-specific antibodies in the context of a bona fide CoV and is a candidate live attenuated SARS-CoV-2 mucosal vaccine that preferentially replicates in the upper airway.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Tests de neutralisation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Humains , Anticorps neutralisants/immunologie , Souris , COVID-19/immunologie , COVID-19/virologie , COVID-19/prévention et contrôle , Anticorps antiviraux/immunologie , Tests de neutralisation/méthodes , Souris transgéniques , Coronavirus humain OC43/immunologie , Coronavirus humain OC43/génétique , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/immunologie , Chlorocebus aethiops , Cellules Vero , Macaca mulatta
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE