Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.539
Filtre
1.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article Dans Anglais | MEDLINE | ID: covidwho-20239015

Résumé

The effective antiviral agents that treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed around the world. The 3C-like protease (3CLpro) of SARS-CoV-2 plays a pivotal role in virus replication; it also has become an important therapeutic target for the infection of SARS-CoV-2. In this work, we have identified Darunavir derivatives that inhibit the 3CLpro through a high-throughput screening method based on a fluorescence resonance energy transfer (FRET) assay in vitro. We found that the compounds 29# and 50# containing polyphenol and caffeine derivatives as the P2 ligand, respectively, exhibited favorable anti-3CLpro potency with EC50 values of 6.3 µM and 3.5 µM and were shown to bind to SARS-CoV-2 3CLpro in vitro. Moreover, we analyzed the binding mode of the DRV in the 3CLpro through molecular docking. Importantly, 29# and 50# exhibited a similar activity against the protease in Omicron variants. The inhibitory effect of compounds 29# and 50# on the SARS-CoV-2 3CLpro warrants that they are worth being the template to design functionally improved inhibitors for the treatment of COVID-19.


Sujets)
Antiviraux , Protéases 3C des coronavirus , Darunavir , Inhibiteurs de protéases , SARS-CoV-2 , Humains , Antiviraux/pharmacologie , COVID-19 , Darunavir/pharmacologie , Simulation de docking moléculaire , Inhibiteurs de protéases/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/enzymologie , Protéases 3C des coronavirus/antagonistes et inhibiteurs
2.
PLoS Pathog ; 18(1): e1010171, 2022 01.
Article Dans Anglais | MEDLINE | ID: covidwho-2327858

Résumé

The development of physiological models that reproduce SARS-CoV-2 infection in primary human cells will be instrumental to identify host-pathogen interactions and potential therapeutics. Here, using cell suspensions directly from primary human lung tissues (HLT), we have developed a rapid platform for the identification of viral targets and the expression of viral entry factors, as well as for the screening of viral entry inhibitors and anti-inflammatory compounds. The direct use of HLT cells, without long-term cell culture and in vitro differentiation approaches, preserves main immune and structural cell populations, including the most susceptible cell targets for SARS-CoV-2; alveolar type II (AT-II) cells, while maintaining the expression of proteins involved in viral infection, such as ACE2, TMPRSS2, CD147 and AXL. Further, antiviral testing of 39 drug candidates reveals a highly reproducible method, suitable for different SARS-CoV-2 variants, and provides the identification of new compounds missed by conventional systems, such as VeroE6. Using this method, we also show that interferons do not modulate ACE2 expression, and that stimulation of local inflammatory responses can be modulated by different compounds with antiviral activity. Overall, we present a relevant and rapid method for the study of SARS-CoV-2.


Sujets)
Antiviraux/usage thérapeutique , , Poumon/virologie , SARS-CoV-2/physiologie , Pénétration virale , Adulte , Animaux , Antiviraux/pharmacologie , COVID-19/immunologie , COVID-19/anatomopathologie , Cellules cultivées , Chlorocebus aethiops , Évaluation préclinique de médicament , Médicaments en essais cliniques/pharmacologie , Médicaments en essais cliniques/usage thérapeutique , Cellules HEK293 , Interactions hôte-pathogène/effets des médicaments et des substances chimiques , Humains , Inflammation/anatomopathologie , Inflammation/thérapie , Inflammation/virologie , Poumon/anatomopathologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Cellules Vero , Pénétration virale/effets des médicaments et des substances chimiques
4.
Appl Environ Microbiol ; 89(6): e0023723, 2023 06 28.
Article Dans Anglais | MEDLINE | ID: covidwho-2317494

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza A virus (IAV), and norovirus are global threats to human health. The application of effective virucidal agents, which contribute to the inactivation of viruses on hands and environmental surfaces, is important to facilitate robust virus infection control measures. Naturally derived virucidal disinfectants have attracted attention owing to their safety and eco-friendly properties. In this study, we showed that multiple Japanese Saxifraga species-derived fractions demonstrated rapid, potent virucidal activity against the SARS-CoV-2 ancestral strain and multiple variant strains, IAV, and two human norovirus surrogates: feline calicivirus (FCV) and murine norovirus (MNV). Condensed tannins were identified as active chemical constituents that play a central role in the virucidal activities of these fractions. At a concentration of 25 µg/mL, the purified condensed tannin fraction Sst-2R induced significant reductions in the viral titers of the SARS-CoV-2 ancestral strain, IAV, and FCV (reductions of ≥3.13, ≥3.00, and 2.50 log10 50% tissue culture infective doses [TCID50]/mL, respectively) within 10 s of reaction time. Furthermore, at a concentration of 100 µg/mL, Sst-2R induced a reduction of 1.75 log10 TCID50/mL in the viral titers of MNV within 1 min. Western blotting and transmission electron microscopy analyses revealed that Sst-2R produced structural abnormalities in viral structural proteins and envelopes, resulting in the destruction of viral particles. Furthermore, Saxifraga species-derived fraction-containing cream showed virucidal activity against multiple viruses within 10 min. Our findings indicate that Saxifraga species-derived fractions containing condensed tannins can be used as disinfectants against multiple viruses on hands and environmental surfaces. IMPORTANCE SARS-CoV-2, IAV, and norovirus are highly contagious pathogens. The use of naturally derived components as novel virucidal/antiviral agents is currently attracting attention. We showed that fractions from extracts of Saxifraga species, in the form of a solution as well as a cream, exerted potent, rapid virucidal activities against SARS-CoV-2, IAV, and surrogates of human norovirus. Condensed tannins were found to play a central role in this activity. The in vitro cytotoxicity of the purified condensed tannin fraction at a concentration that exhibited some extent of virucidal activity was lower than that of 70% ethanol or 2,000 ppm sodium hypochlorite solution, which are popular virucidal disinfectants. Our study suggests that Saxifraga species-derived fractions containing condensed tannins can be used on hands and environmental surfaces as safe virucidal agents against multiple viruses.


Sujets)
Désinfectants , Virus de la grippe A , Norovirus , Proanthocyanidines , SARS-CoV-2 , Saxifragaceae , Désinfectants/pharmacologie , Virus de la grippe A/effets des médicaments et des substances chimiques , Norovirus/effets des médicaments et des substances chimiques , Proanthocyanidines/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Saxifragaceae/composition chimique , Tanins
5.
Viruses ; 14(1)2021 12 27.
Article Dans Anglais | MEDLINE | ID: covidwho-2307471

Résumé

The COVID-19 pandemic has been a public health issue around the world in the last few years. Currently, there is no specific antiviral treatment to fight the disease. Thus, it is essential to highlight possible prognostic predictors that could identify patients with a high risk of developing complications. Within this framework, miRNA biomolecules play a vital role in the genetic regulation of various genes, principally, those related to the pathophysiology of the disease. Here, we review the interaction of host and viral microRNAs with molecular and cellular elements that could potentiate the main pulmonary, cardiac, renal, circulatory, and neuronal complications in COVID-19 patients. miR-26a, miR-29b, miR-21, miR-372, and miR-2392, among others, have been associated with exacerbation of the inflammatory process, increasing the risk of a cytokine storm. In addition, increased expression of miR-15b, -199a, and -491 are related to the prognosis of the disease, and miR-192 and miR-323a were identified as clinical predictors of mortality in patients admitted to the intensive care unit. Finally, we address miR-29, miR-122, miR-155, and miR-200, among others, as possible therapeutic targets. However, more studies are required to confirm these findings.


Sujets)
, COVID-19/diagnostic , microARN/génétique , Antiviraux/administration et posologie , Antiviraux/pharmacologie , COVID-19/complications , COVID-19/génétique , Systèmes de délivrance de médicaments , Interactions hôte-pathogène/effets des médicaments et des substances chimiques , Interactions hôte-pathogène/génétique , Humains , Inflammation , microARN/administration et posologie , Pronostic , ARN viral/génétique , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/génétique
6.
Proc Natl Acad Sci U S A ; 119(32): e2203760119, 2022 08 09.
Article Dans Anglais | MEDLINE | ID: covidwho-2308769

Résumé

The emergence of SARS-CoV-2 variants with enhanced transmissibility, pathogenesis, and resistance to vaccines presents urgent challenges for curbing the COVID-19 pandemic. While Spike mutations that enhance virus infectivity or neutralizing antibody evasion may drive the emergence of these novel variants, studies documenting a critical role for interferon responses in the early control of SARS-CoV-2 infection, combined with the presence of viral genes that limit these responses, suggest that interferons may also influence SARS-CoV-2 evolution. Here, we compared the potency of 17 different human interferons against multiple viral lineages sampled during the course of the global outbreak, including ancestral and five major variants of concern that include the B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma), B.1.617.2 (delta), and B.1.1.529 (omicron) lineages. Our data reveal that relative to ancestral isolates, SARS-CoV-2 variants of concern exhibited increased interferon resistance, suggesting that evasion of innate immunity may be a significant, ongoing driving force for SARS-CoV-2 evolution. These findings have implications for the increased transmissibility and/or lethality of emerging variants and highlight the interferon subtypes that may be most successful in the treatment of early infections.


Sujets)
Antiviraux , COVID-19 , Interférons , SARS-CoV-2 , Anticorps neutralisants , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/transmission , Humains , Interférons/pharmacologie , Interférons/usage thérapeutique , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/génétique
8.
J Biol Chem ; 299(6): 104749, 2023 06.
Article Dans Anglais | MEDLINE | ID: covidwho-2292505

Résumé

The recent SARS-CoV-2 and mpox outbreaks have highlighted the need to expand our arsenal of broad-spectrum antiviral agents for future pandemic preparedness. Host-directed antivirals are an important tool to accomplish this as they typically offer protection against a broader range of viruses than direct-acting antivirals and have a lower susceptibility to viral mutations that cause drug resistance. In this study, we investigate the exchange protein activated by cAMP (EPAC) as a target for broad-spectrum antiviral therapy. We find that the EPAC-selective inhibitor, ESI-09, provides robust protection against a variety of viruses, including SARS-CoV-2 and Vaccinia (VACV)-an orthopox virus from the same family as mpox. We show, using a series of immunofluorescence experiments, that ESI-09 remodels the actin cytoskeleton through Rac1/Cdc42 GTPases and the Arp2/3 complex, impairing internalization of viruses that use clathrin-mediated endocytosis (e.g. VSV) or micropinocytosis (e.g. VACV). Additionally, we find that ESI-09 disrupts syncytia formation and inhibits cell-to-cell transmission of viruses such as measles and VACV. When administered to immune-deficient mice in an intranasal challenge model, ESI-09 protects mice from lethal doses of VACV and prevents formation of pox lesions. Altogether, our finding shows that EPAC antagonists such as ESI-09 are promising candidates for broad-spectrum antiviral therapy that can aid in the fight against ongoing and future viral outbreaks.


Sujets)
Antiviraux , COVID-19 , Orthopoxvirose simienne , Vaccine , Animaux , Souris , Antiviraux/pharmacologie , Orthopoxvirose simienne/traitement médicamenteux , SARS-CoV-2/effets des médicaments et des substances chimiques , Vaccine/traitement médicamenteux , Virus de la vaccine/effets des médicaments et des substances chimiques
9.
Cell ; 185(12): 2086-2102.e22, 2022 06 09.
Article Dans Anglais | MEDLINE | ID: covidwho-2293192

Résumé

Across biological scales, gene-regulatory networks employ autorepression (negative feedback) to maintain homeostasis and minimize failure from aberrant expression. Here, we present a proof of concept that disrupting transcriptional negative feedback dysregulates viral gene expression to therapeutically inhibit replication and confers a high evolutionary barrier to resistance. We find that nucleic-acid decoys mimicking cis-regulatory sites act as "feedback disruptors," break homeostasis, and increase viral transcription factors to cytotoxic levels (termed "open-loop lethality"). Feedback disruptors against herpesviruses reduced viral replication >2-logs without activating innate immunity, showed sub-nM IC50, synergized with standard-of-care antivirals, and inhibited virus replication in mice. In contrast to approved antivirals where resistance rapidly emerged, no feedback-disruptor escape mutants evolved in long-term cultures. For SARS-CoV-2, disruption of a putative feedback circuit also generated open-loop lethality, reducing viral titers by >1-log. These results demonstrate that generating open-loop lethality, via negative-feedback disruption, may yield a class of antimicrobials with a high genetic barrier to resistance.


Sujets)
Antiviraux , Régulation de l'expression des gènes viraux/effets des médicaments et des substances chimiques , Animaux , Antiviraux/pharmacologie , Résistance virale aux médicaments , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Souris , SARS-CoV-2/effets des médicaments et des substances chimiques , Réplication virale
10.
Curr Opin Pulm Med ; 27(3): 169-175, 2021 05 01.
Article Dans Anglais | MEDLINE | ID: covidwho-2286794

Résumé

PURPOSE OF REVIEW: Coronavirus disease 2019 (COVID-19) is an acute multisystem disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Investigations are ongoing in the search for effective therapeutics, with clinical approaches evolving based upon such evidence. RECENT FINDINGS: The antiviral agent, remdesivir, and the immunomodulator, dexamethasone, are the first therapeutics for which there is evidence of efficacy from randomized trials. Subgroup analyses suggest remdesivir is beneficial in hospitalized patients whose severity of illness falls at the lower end of the spectrum, while dexamethasone is more beneficial in hospitalized patients whose severity of illness falls at the higher end of the spectrum. We recommend that inpatients who require supplemental oxygen but are not mechanically ventilated receive both remdesivir and dexamethasone, and inpatients who require mechanical ventilation receive dexamethasone monotherapy. Additional evidence regarding anti-SARS-CoV-2 antibodies, convalescent plasma, and a variety of antiinterleukin therapies is forthcoming. SUMMARY: The body of evidence related to COVID-19 therapeutics continues to evolve and, as a result, management is likely to change with time. As new evidence is generated and published, the optimal approach to managing patients with COVID-19 should be reconsidered.


Sujets)
AMP/analogues et dérivés , Alanine/analogues et dérivés , COVID-19/thérapie , Dexaméthasone/pharmacologie , Ventilation artificielle/méthodes , AMP/pharmacologie , Alanine/pharmacologie , Antiviraux/pharmacologie , COVID-19/immunologie , Humains , Immunisation passive/méthodes , Facteurs immunologiques/pharmacologie , Sélection de patients , SARS-CoV-2/effets des médicaments et des substances chimiques ,
11.
Lett Appl Microbiol ; 71(3): 229-234, 2020 Sep.
Article Dans Anglais | MEDLINE | ID: covidwho-2248530

Résumé

Understanding mechanisms of the novel SARS-CoV2 infection and progression can provide potential novel targets for prevention and/or treatment. This could be achieved via the inhibition of viral entry and/or replication, or by suppression of the immunologic response that is provoked by the infection (known as the cytokine storm). Probiotics are defined as 'live microorganisms that, when administered in adequate amounts, confer a health benefit on the host'. There is scarcity of evidence about the relationship between COVID-19 and gut microbiota. So, whether or not these supplements can prevent or ameliorate COVID-19-associated symptoms is not fully understood. The aim of this study is to provide an indirect evidence about the utility of probiotics in combating COVID-19 or its associated symptoms, through the review of its antiviral and anti-inflammatory properties in vitro, animal models and human trials. SIGNIFICANCE AND IMPACT OF THE STUDY: The role of probiotics in alleviation of the novel COVID-19 has not been established. This review provides an insight about the anti-inflammatory, antiviral effects of probiotics in vitro, animal models and human. The latter can provide an indirect evidence and/or hypothesis-driven approach to investigate the use of probiotics as adjunctive therapy in the prophylaxis and/or alleviation of COVID-19 symptoms.


Sujets)
Anti-inflammatoires/usage thérapeutique , COVID-19/diétothérapie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Probiotiques/usage thérapeutique , SARS-CoV-2/effets des médicaments et des substances chimiques , Animaux , Cytokines/sang , Compléments alimentaires , Humains , Rats
12.
Signal Transduct Target Ther ; 8(1): 128, 2023 03 16.
Article Dans Anglais | MEDLINE | ID: covidwho-2263420

Résumé

Emerging SARS-CoV-2 variants, particularly the Omicron variant and its sublineages, continually threaten the global public health. Small molecule antivirals are an effective treatment strategy to fight against the virus. However, the first-generation antivirals either show limited clinical efficacy and/or have some defects in pharmacokinetic (PK) properties. Moreover, with increased use of these drugs across the globe, they face great pressure of drug resistance. We herein present the discovery and characterization of a new generation antiviral drug candidate (SY110), which is a potent and selective inhibitor of SARS-CoV-2 main protease (Mpro). This compound displayed potent in vitro antiviral activity against not only the predominant SARS-CoV-2 Omicron sublineage BA.5, but also other highly pathogenic human coronaviruses including SARS-CoV-1 and MERS-CoV. In the Omicron-infected K18-hACE2 mouse model, oral treatment with SY110 significantly lowered the viral burdens in lung and alleviated the virus-induced pathology. Importantly, SY110 possesses favorable PK properties with high oral drug exposure and oral bioavailability, and also an outstanding safety profile. Furthermore, SY110 exhibited sensitivity to several drug-resistance Mpro mutations. Collectively, this investigation provides a promising new drug candidate against Omicron and other variants of SARS-CoV-2.


Sujets)
COVID-19 , Protéases 3C des coronavirus , SARS-CoV-2 , Animaux , Humains , Souris , Administration par voie orale , Antiviraux/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/métabolisme , /méthodes , Protéases 3C des coronavirus/antagonistes et inhibiteurs
13.
Nat Commun ; 14(1): 1076, 2023 02 25.
Article Dans Anglais | MEDLINE | ID: covidwho-2262859

Résumé

COVID-19 caused by SARS-CoV-2 has continually been serious threat to public health worldwide. While a few anti-SARS-CoV-2 therapeutics are currently available, their antiviral potency is not sufficient. Here, we identify two orally available 4-fluoro-benzothiazole-containing small molecules, TKB245 and TKB248, which specifically inhibit the enzymatic activity of main protease (Mpro) of SARS-CoV-2 and significantly more potently block the infectivity and replication of various SARS-CoV-2 strains than nirmatrelvir, molnupiravir, and ensitrelvir in cell-based assays employing various target cells. Both compounds also block the replication of Delta and Omicron variants in human-ACE2-knocked-in mice. Native mass spectrometric analysis reveals that both compounds bind to dimer Mpro, apparently promoting Mpro dimerization. X-ray crystallographic analysis shows that both compounds bind to Mpro's active-site cavity, forming a covalent bond with the catalytic amino acid Cys-145 with the 4-fluorine of the benzothiazole moiety pointed to solvent. The data suggest that TKB245 and TKB248 might serve as potential therapeutics for COVID-19 and shed light upon further optimization to develop more potent and safer anti-SARS-CoV-2 therapeutics.


Sujets)
Antiviraux , COVID-19 , Protéases 3C des coronavirus , Inhibiteurs de protéases , SARS-CoV-2 , Animaux , Humains , Souris , Antiviraux/pharmacologie , Benzothiazoles , Simulation de docking moléculaire , Inhibiteurs de protéases/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Protéines virales non structurales/composition chimique , Protéases 3C des coronavirus/antagonistes et inhibiteurs
14.
Int J Mol Sci ; 24(5)2023 Feb 28.
Article Dans Anglais | MEDLINE | ID: covidwho-2267326

Résumé

We report synthesis of a novel 1,2,3,4-tetrahydroquinazoline derivative, named 2-(6,8-dibromo-3-(4-hydroxycyclohexyl)-1,2,3,4-tetrahydroquinazolin-2-yl)phenol (1), which was obtained from the hydrochloride of 4-((2-amino-3,5-dibromobenzyl)amino)cyclohexan-1-ol (ambroxol hydrochloride) and salicylaldehyde in EtOH. The resulting compound was produced in the form of colorless crystals of the composition 1∙0.5EtOH. The formation of the single product was confirmed by the IR and 1H spectroscopy, single-crystal and powder X-ray diffraction, and elemental analysis. The molecule of 1 contains a chiral tertiary carbon of the 1,2,3,4-tetrahydropyrimidine fragment and the crystal structure of 1∙0.5EtOH is a racemate. Optical properties of 1∙0.5EtOH were revealed by UV-vis spectroscopy in MeOH and it was established that the compound absorbs exclusively in the UV region up to about 350 nm. 1∙0.5EtOH in MeOH exhibits dual emission and the emission spectra contains bands at about 340 and 446 nm upon excitation at 300 and 360 nm, respectively. The DFT calculations were performed to verify the structure as well as electronic and optical properties of 1. ADMET properties of the R-isomer of 1 were evaluated using the SwissADME, BOILED-Egg, and ProTox-II tools. As evidenced from the blue dot position in the BOILED-Egg plot, both human blood-brain barrier penetration and gastrointestinal absorption properties are positive with the positive PGP effect on the molecule. Molecular docking was applied to examine the influence of the structures of both R-isomer and S-isomer of 1 on a series of the SARS-CoV-2 proteins. According to the docking analysis results, both isomers of 1 were found to be active against all the applied SARS-CoV-2 proteins with the best binding affinities with Papain-like protease (PLpro) and nonstructural protein 3 (Nsp3_range 207-379-AMP). Ligand efficiency scores for both isomers of 1 inside the binding sites of the applied proteins were also revealed and compared with the initial ligands. Molecular dynamics simulations were also applied to evaluate the stability of complexes of both isomers with Papain-like protease (PLpro) and nonstructural protein 3 (Nsp3_range 207-379-AMP). The complex of the S-isomer with Papain-like protease (PLpro) was found to be highly unstable, while the other complexes are stable.


Sujets)
Ambroxol , COVID-19 , Protéases de type papaïne des coronavirus , Quinazolines , SARS-CoV-2 , Humains , Ambroxol/analogues et dérivés , Ambroxol/pharmacocinétique , Ambroxol/pharmacologie , Simulation de docking moléculaire , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/enzymologie , Quinazolines/composition chimique , Quinazolines/pharmacocinétique , Quinazolines/pharmacologie , Protéases de type papaïne des coronavirus/antagonistes et inhibiteurs , Protéases de type papaïne des coronavirus/composition chimique
15.
J Med Chem ; 66(4): 3088-3105, 2023 02 23.
Article Dans Anglais | MEDLINE | ID: covidwho-2265584

Résumé

Interest in covalent enzyme inhibitors as therapeutic agents has seen a recent resurgence. Covalent enzyme inhibitors typically possess an organic functional group that reacts with a key feature of the target enzyme, often a nucleophilic cysteine residue. Herein, the application of small, modular ReV complexes as inorganic cysteine-targeting warheads is described. These metal complexes were found to react with cysteine residues rapidly and selectively. To demonstrate the utility of these ReV complexes, their reactivity with SARS-CoV-2-associated cysteine proteases is presented, including the SARS-CoV-2 main protease and papain-like protease and human enzymes cathepsin B and L. As all of these proteins are cysteine proteases, these enzymes were found to be inhibited by the ReV complexes through the formation of adducts. These findings suggest that these ReV complexes could be used as a new class of warheads for targeting surface accessible cysteine residues in disease-relevant target proteins.


Sujets)
COVID-19 , Cysteine proteases , Inhibiteurs de la cystéine protéinase , Cystéine , Rhénium , SARS-CoV-2 , Humains , Cysteine proteases/métabolisme , Antienzymes , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/enzymologie , Inhibiteurs de la cystéine protéinase/composition chimique , Inhibiteurs de la cystéine protéinase/pharmacologie , Inhibiteurs de la cystéine protéinase/usage thérapeutique
16.
Cell Biochem Funct ; 41(3): 284-295, 2023 Apr.
Article Dans Anglais | MEDLINE | ID: covidwho-2275936

Résumé

In the last 2 years, different pharmacological agents have been indicated as potential inhibitors of SARS-CoV-2 in vitro. Specifically, drugs termed as functional inhibitors of acid sphingomyelinase (FIASMAs) have proved to inhibit the SARS-CoV-2 replication using different types of cells. Those therapeutic agents share several chemical structure characteristics and some well-known representatives are fluoxetine, escitalopram, fluvoxamine, and others. Most of the FIASMAs are primarily used as effective therapeutic agents to treat different pathologies, therefore, they are natural drug candidates for repositioning strategy. In this review, we summarize the two main proposed mechanisms mediating acid sphingomyelinase (ASM) inhibition and how they can explain the inhibition of SARS-CoV-2 replication by FIASMAs. The first mechanism implies a disruption in the lysosomal pH fall as the endosome-lysosome moves toward the interior of the cell. In fact, changes in cholesterol levels in endosome-lysosome membranes, which are associated with ASM inhibition is thought to be mediated by lysosomal proton pump (ATP-ase) inactivation. The second mechanism involves the formation of an extracellular ceramide-rich domain, which is blocked by FIASMAs. The ceramide-rich domains are believed to facilitate the SARS-CoV-2 entrance into the host cells.


Sujets)
COVID-19 , SARS-CoV-2 , Sphingomyeline phosphodiesterase , Humains , Céramides/métabolisme , Fluoxétine/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Sphingomyeline phosphodiesterase/antagonistes et inhibiteurs , Sphingomyeline phosphodiesterase/métabolisme
17.
Nucleic Acids Res ; 51(9): 4555-4571, 2023 05 22.
Article Dans Anglais | MEDLINE | ID: covidwho-2275338

Résumé

The pandemic caused by SARS-CoV-2 has called for concerted efforts to generate new insights into the biology of betacoronaviruses to inform drug screening and development. Here, we establish a workflow to determine the RNA recognition and druggability of the nucleocapsid N-protein of SARS-CoV-2, a highly abundant protein crucial for the viral life cycle. We use a synergistic method that combines NMR spectroscopy and protein-RNA cross-linking coupled to mass spectrometry to quickly determine the RNA binding of two RNA recognition domains of the N-protein. Finally, we explore the druggability of these domains by performing an NMR fragment screening. This workflow identified small molecule chemotypes that bind to RNA binding interfaces and that have promising properties for further fragment expansion and drug development.


Sujets)
, COVID-19 , Protéines de la nucléocapside des coronavirus , Développement de médicament , SARS-CoV-2 , Humains , COVID-19/virologie , ARN viral/métabolisme , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/métabolisme , Protéines de la nucléocapside des coronavirus/antagonistes et inhibiteurs , Protéines de la nucléocapside des coronavirus/composition chimique , Protéines de la nucléocapside des coronavirus/métabolisme , Résonance magnétique nucléaire biomoléculaire , Spectrométrie de masse , Flux de travaux , Liaison aux protéines
18.
Int J Mol Sci ; 24(4)2023 Feb 20.
Article Dans Anglais | MEDLINE | ID: covidwho-2281048

Résumé

The indispensable role of the SARS-CoV-2 main protease (Mpro) in the viral replication cycle and its dissimilarity to human proteases make Mpro a promising drug target. In order to identify the non-covalent Mpro inhibitors, we performed a comprehensive study using a combined computational strategy. We first screened the ZINC purchasable compound database using the pharmacophore model generated from the reference crystal structure of Mpro complexed with the inhibitor ML188. The hit compounds were then filtered by molecular docking and predicted parameters of drug-likeness and pharmacokinetics. The final molecular dynamics (MD) simulations identified three effective candidate inhibitors (ECIs) capable of maintaining binding within the substrate-binding cavity of Mpro. We further performed comparative analyses of the reference and effective complexes in terms of dynamics, thermodynamics, binding free energy (BFE), and interaction energies and modes. The results reveal that, when compared to the inter-molecular electrostatic forces/interactions, the inter-molecular van der Waals (vdW) forces/interactions are far more important in maintaining the association and determining the high affinity. Given the un-favorable effects of the inter-molecular electrostatic interactions-association destabilization by the competitive hydrogen bond (HB) interactions and the reduced binding affinity arising from the un-compensable increase in the electrostatic desolvation penalty-we suggest that enhancing the inter-molecular vdW interactions while avoiding introducing the deeply buried HBs may be a promising strategy in future inhibitor optimization.


Sujets)
Protéases 3C des coronavirus , Inhibiteurs de protéases , SARS-CoV-2 , Humains , COVID-19 , Simulation de docking moléculaire , SARS-CoV-2/effets des médicaments et des substances chimiques , Protéases 3C des coronavirus/antagonistes et inhibiteurs
19.
ACS Biomater Sci Eng ; 9(3): 1656-1671, 2023 03 13.
Article Dans Anglais | MEDLINE | ID: covidwho-2271527

Résumé

As the world braces to enter its fourth year of the coronavirus disease 2019 (COVID-19) pandemic, the need for accessible and effective antiviral therapeutics continues to be felt globally. The recent surge of Omicron variant cases has demonstrated that vaccination and prevention alone cannot quell the spread of highly transmissible variants. A safe and nontoxic therapeutic with an adaptable design to respond to the emergence of new variants is critical for transitioning to the treatment of COVID-19 as an endemic disease. Here, we present a novel compound, called SBCoV202, that specifically and tightly binds the translation initiation site of RNA-dependent RNA polymerase within the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome, inhibiting viral replication. SBCoV202 is a Nanoligomer, a molecule that includes peptide nucleic acid sequences capable of binding viral RNA with single-base-pair specificity to accurately target the viral genome. The compound has been shown to be safe and nontoxic in mice, with favorable biodistribution, and has shown efficacy against SARS-CoV-2 in vitro. Safety and biodistribution were assessed using three separate administration methods, namely, intranasal, intravenous, and intraperitoneal. Safety studies showed the Nanoligomer caused no outward distress, immunogenicity, or organ tissue damage, measured through observation of behavior and body weight, serum levels of cytokines, and histopathology of fixed tissue, respectively. SBCoV202 was evenly biodistributed throughout the body, with most tissues measuring Nanoligomer concentrations well above the compound KD of 3.37 nM. In addition to favorable availability to organs such as the lungs, lymph nodes, liver, and spleen, the compound circulated through the blood and was rapidly cleared through the renal and urinary systems. The favorable biodistribution and lack of immunogenicity and toxicity set Nanoligomers apart from other antisense therapies, while the adaptability of the nucleic acid sequence of Nanoligomers provides a defense against future emergence of drug resistance, making these molecules an attractive potential treatment for COVID-19.


Sujets)
, COVID-19 , Génome viral , Nanomédecine , Nanostructures , Oligoribonucléotides , Acides nucléiques peptidiques , SARS-CoV-2 , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/génétique , COVID-19/virologie , /effets indésirables , /méthodes , Nanostructures/administration et posologie , Nanostructures/effets indésirables , Nanostructures/usage thérapeutique , Nanomédecine/méthodes , Sécurité des patients , Acides nucléiques peptidiques/administration et posologie , Acides nucléiques peptidiques/effets indésirables , Acides nucléiques peptidiques/pharmacocinétique , Acides nucléiques peptidiques/usage thérapeutique , Oligoribonucléotides/administration et posologie , Oligoribonucléotides/effets indésirables , Oligoribonucléotides/pharmacocinétique , Oligoribonucléotides/usage thérapeutique , Animaux , Souris , Souris de lignée BALB C , Techniques in vitro , Génome viral/effets des médicaments et des substances chimiques , Génome viral/génétique , Distribution tissulaire
20.
J Med Chem ; 65(4): 2747-2784, 2022 02 24.
Article Dans Anglais | MEDLINE | ID: covidwho-2275124

Résumé

Analysis of the SARS-CoV-2 sequence revealed a multibasic furin cleavage site at the S1/S2 boundary of the spike protein distinguishing this virus from SARS-CoV. Furin, the best-characterized member of the mammalian proprotein convertases, is an ubiquitously expressed single pass type 1 transmembrane protein. Cleavage of SARS-CoV-2 spike protein by furin promotes viral entry into lung cells. While furin knockout is embryonically lethal, its knockout in differentiated somatic cells is not, thus furin provides an exciting therapeutic target for viral pathogens including SARS-CoV-2 and bacterial infections. Several peptide-based and small-molecule inhibitors of furin have been recently reported, and select cocrystal structures have been solved, paving the way for further optimization and selection of clinical candidates. This perspective highlights furin structure, substrates, recent inhibitors, and crystal structures with emphasis on furin's role in SARS-CoV-2 infection, where the current data strongly suggest its inhibition as a promising therapeutic intervention for SARS-CoV-2.


Sujets)
Antiviraux/pharmacologie , Furine/antagonistes et inhibiteurs , Peptides/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/pharmacologie , Glycoprotéine de spicule des coronavirus/antagonistes et inhibiteurs , Animaux , Antiviraux/composition chimique , COVID-19/métabolisme , Furine/métabolisme , Humains , Peptides/composition chimique , SARS-CoV-2/métabolisme , Bibliothèques de petites molécules/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme ,
SÉLECTION CITATIONS
Détails de la recherche