Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sustainable Materials and Technologies ; 35, 2023.
Article in English | Scopus | ID: covidwho-2245255

ABSTRACT

The rapid transmission of contagious viruses responsible for global pandemic and various extraordinary risk to precious human life including death. For instance, the current ongoing worldwide COVID-19 pandemic caused by novel coronavirus (SARS-CoV-2) is a communicable disease which is transmitted via touching the contaminated surfaces and then nosocomial route. In absence of effective vaccines and therapies, antiviral coatings are essential in order to prevent or slowdown rapid transmission of viruses. In this prospective, sustainable nanotechnology and material engineering have provided substantial contribution in development of engineered nanomaterial based antiviral coated surfaces to the humanity. In the recent past, nanomaterials based on silver (Ag), titanium oxide (TiO2), copper sulfide (CuS) and copper oxide (CuO) have been modified in the form of engineered nanomaterials with effective antiviral efficacy against SARS-CoV-2. In this review, various recent fundamental aspects for fabrication of metallic nanoparticles (Ag, Ti, Cu etc.) based coated surfaces on various substrates and their antiviral efficacy to inhibit viral transmission of SARS-CoV-2 are discussed along with their respective conceptual mechanisms. The antiviral mechanism based on chemistry of engineered nanomaterials is the key outcome of this review that would be useful for future research in designing and development of more advance antiviral materials and coated surfaces in order to control of future epidemics. © 2022 Elsevier B.V.

2.
Antiviral Res ; 209: 105465, 2022 Nov 17.
Article in English | MEDLINE | ID: covidwho-2231152

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak has resulted in significant global morbidity, mortality, and societal disruption. Currently, effective antiviral drugs for the treatment of SARS-CoV-2 infection are limited. Therefore, safe and effective antiviral drugs to combat COVID-19 are urgently required. In previous studies, we showed that 3-indoleacetonitrile, a plant growth hormone produced by cruciferous (Brassica) vegetables, is effective in treating influenza A virus infection. However, the molecular mechanisms underlying these effects remain unclear. Herein, we demonstrated that 3-indoleacetonitrile exhibits broad-spectrum antiviral activity and is effective against HSV-1 and VSV infections in vitro. This phenomenon prompted us to study its role in the anti-SARS-CoV-2 process. Interestingly, 3-indoleacetonitrile exhibited antiviral activity against SARS-CoV-2 in vitro. Importantly, tail vein injection of 3-indoleacetonitrile resulted in good antiviral activity in mouse models infected with WBP-1 (a mouse adaptation of the SARS-CoV-2 strain). Mechanistically, 3-indoleacetonitrile promoted the host interferon signalling pathway response and inhibited autophagic flux. Furthermore, we demonstrated that 3-indoleacetonitrile induced an increase in mitochondrial antiviral-signalling (MAVS) protein levels, which might be attributed to its inhibition of the interaction between MAVS and the selective autophagy receptor SQSTM1. Overall, our results demonstrate that 3-indoleacetonitrile is potently active against SARS-CoV-2 in vitro and in vivo, which may provide a foundation for further clinical testing for the treatment of COVID-19. In addition, considering its broad-spectrum antiviral effect, it should be explored whether it also has an effect on other viruses that threaten human health.

3.
MedComm (2020) ; 4(1): e206, 2023 Feb.
Article in English | MEDLINE | ID: covidwho-2209138

ABSTRACT

Tetrandrine (TET) has been used to treat silicosis in China for decades. The aim of this study was to facilitate rational repurposing of TET against SARS-CoV-2 infection. In this study, we confirmed that TET exhibited antiviral potency against SARS-CoV-2 in the African green monkey kidney (Vero E6), human hepatocarcinoma (Huh7), and human lung adenocarcinoma epithelial (Calu-3) cell lines. TET functioned during the early-entry stage of SARS-CoV-2 and impeded intracellular trafficking of the virus from early endosomes to endolysosomes. An in vivo study that used adenovirus (AdV) 5-human angiotensin-converting enzyme 2 (hACE2)-transduced mice showed that although TET did not reduce pulmonary viral load, it significantly alleviated pathological damage in SARS-CoV-2-infected murine lungs. The systemic preclinical pharmacokinetics were investigated based on in vivo and in vitro models, and the route-dependent biodistribution of TET was explored. TET had a large volume of distribution, which contributed to its high tissue accumulation. Inhaled administration helped TET target the lung and reduced its exposure to other tissues, which mitigated its off-target toxicity. Based on the available human pharmacokinetic data, it appeared feasible to achieve an unbound TET 90% maximal effective concentration (EC90) in human lungs. This study provides insights into the route-dependent pulmonary biodistribution of TET associated with its efficacy.

4.
Sustainable Materials and Technologies ; : e00544, 2022.
Article in English | ScienceDirect | ID: covidwho-2150642

ABSTRACT

The rapid transmission of contagious viruses responsible for global pandemic and various extraordinary risk to precious human life including death. For instance, the current ongoing worldwide COVID-19 pandemic caused by novel coronavirus (SARS-CoV-2) is a communicable disease which is transmitted via touching the contaminated surfaces and then nosocomial route. In absence of effective vaccines and therapies, antiviral coatings are essential in order to prevent or slowdown rapid transmission of viruses. In this prospective, sustainable nanotechnology and material engineering have provided substantial contribution in development of engineered nanomaterial based antiviral coated surfaces to the humanity. In the recent past, nanomaterials based on silver (Ag), titanium oxide (TiO2), copper sulfide (CuS) and copper oxide (CuO) have been modified in the form of engineered nanomaterials with effective antiviral efficacy against SARS-CoV-2. In this review, various recent fundamental aspects for fabrication of metallic nanoparticles (Ag, Ti, Cu etc.) based coated surfaces on various substrates and their antiviral efficacy to inhibit viral transmission of SARS-CoV-2 are discussed along with their respective conceptual mechanisms. The antiviral mechanism based on chemistry of engineered nanomaterials is the key outcome of this review that would be useful for future research in designing and development of more advance antiviral materials and coated surfaces in order to control of future epidemics.

5.
Front Pharmacol ; 13: 939573, 2022.
Article in English | MEDLINE | ID: covidwho-1928445

ABSTRACT

Background: The rapid worldwide spread of the Omicron variant of SARS-CoV-2 has unleashed a new wave of COVID-19 outbreaks. The efficacy of molnupiravir, an approved drug, is still unknown in patients infected with the Omicron variant. Objective: Evaluated the antiviral efficacy and safety of molnupiravir in patients infected with SARS-CoV-2 Omicron variant, with symptom duration within 5 days. Methods: We conducted a randomized, controlled trial involving patients with mild or moderate COVID-19. Patients were randomized to orally receive molnupiravir (800 mg) plus basic treatment or only basic treatment for 5 days (BID). The antiviral efficacy of the drug was evaluated using reverse transcriptase polymerase chain reaction. Results: Results showed that the time of viral RNA clearance (primary endpoint) was significantly decreased in the molnupiravir group (median, 9 days) compared to the control group (median, 10 days) (Log-Rank p = 0.0092). Of patients receiving molnupiravir, 18.42% achieved viral RNA clearance on day 5 of treatment, compared to the control group (0%) (p = 0.0092). On day 7, 40.79%, and 6.45% of patients in the molnupiravir and control groups, respectively, achieved viral RNA clearance (p = 0.0004). In addition, molnupiravir has a good safety profile, and no serious adverse events were reported. Conclusion: Molnupiravir significantly accelerated the SARS-CoV-2 Omicron RNA clearance in patients with COVID-19. Clinical Trial Registration: [chictr.org.cn], identifier [ChiCTR2200056817].

6.
Viruses ; 14(4)2022 04 06.
Article in English | MEDLINE | ID: covidwho-1776362

ABSTRACT

The ongoing global pandemic of COVID-19 poses unprecedented public health risks for governments and societies around the world, which have been exacerbated by the emergence of SARS-CoV-2 variants. Pharmaceutical interventions with high antiviral efficacy are expected to delay and contain the COVID-19 pandemic. Molnupiravir, as an oral antiviral prodrug, is active against SARS-CoV-2 and is now (23 February 2022) one of the seven widely-used coronavirus treatments. To estimate its antiviral efficacy of Molnupiravir, we built a granular mathematical within-host model. We find that the antiviral efficacy of Molnupiravir to stop the growth of the virus is 0.56 (95% CI: 0.49, 0.64), which could inhibit 56% of the replication of infected cells per day. There has been good progress in developing high-efficacy antiviral drugs that rapidly reduce viral load and may also reduce the infectiousness of treated cases if administered as early as possible.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cytidine/analogs & derivatives , Humans , Hydroxylamines , Pandemics
7.
Viruses ; 14(1)2021 12 24.
Article in English | MEDLINE | ID: covidwho-1580407

ABSTRACT

Only a mere fraction of the huge variety of human pathogenic viruses can be targeted by the currently available spectrum of antiviral drugs. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak has highlighted the urgent need for molecules that can be deployed quickly to treat novel, developing or re-emerging viral infections. Sulfated polysaccharides are found on the surfaces of both the susceptible host cells and the majority of human viruses, and thus can play an important role during viral infection. Such polysaccharides widely occurring in natural sources, specifically those converted into sulfated varieties, have already proved to possess a high level and sometimes also broad-spectrum antiviral activity. This antiviral potency can be determined through multifold molecular pathways, which in many cases have low profiles of cytotoxicity. Consequently, several new polysaccharide-derived drugs are currently being investigated in clinical settings. We reviewed the present status of research on sulfated polysaccharide-based antiviral agents, their structural characteristics, structure-activity relationships, and the potential of clinical application. Furthermore, the molecular mechanisms of sulfated polysaccharides involved in viral infection or in antiviral activity, respectively, are discussed, together with a focus on the emerging methodology contributing to polysaccharide-based drug development.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , COVID-19/epidemiology , Polysaccharides/pharmacology , Viruses/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Heparin/chemical synthesis , Heparin/chemistry , Heparin/pharmacology , Humans , Polysaccharides/chemistry , SARS-CoV-2/drug effects , Structure-Activity Relationship , Sulfates/chemistry , Sulfates/pharmacology , Virus Diseases/drug therapy , Virus Internalization/drug effects , Viruses/pathogenicity , COVID-19 Drug Treatment
8.
Int J Antimicrob Agents ; 59(1): 106499, 2022 Jan.
Article in English | MEDLINE | ID: covidwho-1587677

ABSTRACT

In a bid to contain the current COVID-19 (coronavirus disease 2019) pandemic, various countermeasures have been applied. To date, however, there is a lack of an effective drug for the treatment of COVID-19. Through molecular modelling studies, simeprevir, a protease inhibitor approved for the management of hepatitis C virus infection, has been predicted as a potential antiviral against SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), the causative agent of COVID-19. Here we assessed the efficacy of simeprevir against SARS-CoV-2 both in vitro in Vero E6 cells and in vivo in a human angiotensin-converting enzyme 2 (hACE2) transgenic mouse model. The results showed that simeprevir could inhibit SARS-CoV-2 replication in Vero E6 cells with a half-maximal effective concentration (EC50) of 1.41 ± 0.12 µM. In a transgenic hACE2 mouse model of SARS-CoV-2 infection, intraperitoneal administration of simeprevir at 10 mg/kg/day for 3 consecutive days failed to suppress viral replication. These findings collectively imply that simeprevir does not inhibit SARS-CoV-2 in vivo and therefore do not support its application as a treatment against COVID-19 at a dosage of 10 mg/kg/day.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Antiviral Agents/pharmacology , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , Simeprevir/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/therapeutic use , COVID-19/virology , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Transgenic , Negative Results , Protease Inhibitors/therapeutic use , Simeprevir/therapeutic use , Vero Cells , COVID-19 Drug Treatment
9.
Viruses ; 13(8)2021 08 23.
Article in English | MEDLINE | ID: covidwho-1524167

ABSTRACT

The pandemic of COVID-19 caused by SARS-CoV-2 continues to spread despite the global efforts taken to control it. The 3C-like protease (3CLpro), the major protease of SARS-CoV-2, is one of the most interesting targets for antiviral drug development because it is highly conserved among SARS-CoVs and plays an important role in viral replication. Herein, we developed high throughput screening for SARS-CoV-2 3CLpro inhibitor based on AlphaScreen. We screened 91 natural product compounds and found that all-trans retinoic acid (ATRA), an FDA-approved drug, inhibited 3CLpro activity. The 3CLpro inhibitory effect of ATRA was confirmed in vitro by both immunoblotting and AlphaScreen with a 50% inhibition concentration (IC50) of 24.7 ± 1.65 µM. ATRA inhibited the replication of SARS-CoV-2 in VeroE6/TMPRSS2 and Calu-3 cells, with IC50 = 2.69 ± 0.09 µM in the former and 0.82 ± 0.01 µM in the latter. Further, we showed the anti-SARS-CoV-2 effect of ATRA on the currently circulating variants of concern (VOC); alpha, beta, gamma, and delta. These results suggest that ATRA may be considered as a potential therapeutic agent against SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , SARS-CoV-2/drug effects , Tretinoin/pharmacology , Animals , Cell Line, Tumor , Chlorocebus aethiops , Cysteine Proteinase Inhibitors/pharmacology , DEAD Box Protein 58/metabolism , High-Throughput Screening Assays , Humans , Receptors, Immunologic/metabolism , SARS-CoV-2/enzymology , SARS-CoV-2/physiology , Vero Cells , Virus Replication/drug effects
10.
J Adv Vet Anim Res ; 8(1): 24-35, 2021 Mar.
Article in English | MEDLINE | ID: covidwho-1190580

ABSTRACT

OBJECTIVES: The comprehensive in silico study aims to figure out the most effective aromatic phytochemical ligands among a number from a library, considering their pharmacokinetic efficacies in blocking "angiotensin-converting enzyme 2 (ACE2) receptor-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) S protein" complex formation as part of a target-specific drug designing. MATERIALS AND METHODS: A library of 57 aromatic pharmacophore phytochemical ligands was prepared from where the top five ligands depending on Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) and quantitative structure-activity relationship (QSAR)-based pharmacokinetic properties were considered. The selected ligands were optimized for commencing molecular docking and dynamic simulation as a complex with the ACE2 receptor to compare their blocking efficacy with the control drug. The ligand-receptor complexes' accuracy in preventing the Spike (S) protein of SARS-CoV-2 penetration inside the host cells has been analyzed through hydrogen-hydrophobic bond interactions, principal component analysis (PCA), root mean square deviation (RMSD), root mean square fluctuation (RMSF), and B-Factor. Advanced in silico programming language and bioanalytical software were used for high throughput and authentic results. RESULTS: ADMET and QSAR revealed Rhamnetin, Lactupicrin, Rhinacanthin D, Flemiflavanone D, and Exiguaflavanone A as the ligands of our interest to be compared with the control Cassiarin D. According to the molecular docking binding affinity to block ACE2 receptor, the efficiency mountings were Rhinacanthin D > Flemiflavanone D > Lactupicrin > Exiguaflavanone A > Rhamnetin. The binding affinity of the Cassiarin D-ACE2 complex was (-10.2 KJ/mol) found inferior to the Rhinacanthin D-ACE2 complex (-10.8 KJ/mol), referring to Rhinacanthin D as a more stable candidate to use as drugs. The RMSD values of protein-ligand complexes evaluated according to their structural conformation and stable binding pose ranged between 0.1~2.1 Å. The B-factor showed that very few loops were present in the protein structure. The RMSF peak fluctuation regions ranged 5-250, predicting efficient ligand-receptor interactions. CONCLUSION: The experiment sequentially measures all the parameters required in referring to any pharmacophore as a drug, considering which all aromatic components analyzed in the study can strongly be predicted as target-specific medication against the novel coronavirus 2019 infection.

11.
Antiviral Res ; 189: 105056, 2021 05.
Article in English | MEDLINE | ID: covidwho-1126675

ABSTRACT

Emetine is a FDA-approved drug for the treatment of amebiasis. Previously we demonstrated the antiviral efficacy of emetine against some RNA and DNA viruses. In this study, we evaluated the in vitro antiviral efficacy of emetine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and found it to be a low nanomolar (nM) inhibitor. Interestingly, emetine exhibited protective efficacy against lethal challenge with infectious bronchitis virus (IBV; a chicken coronavirus) in the embryonated chicken egg infection model. Emetine treatment led to a decrease in viral RNA and protein synthesis without affecting other steps of viral life cycle such as attachment, entry and budding. In a chromatin immunoprecipitation (CHIP) assay, emetine was shown to disrupt the binding of SARS-CoV-2 mRNA with eIF4E (eukaryotic translation initiation factor 4E, a cellular cap-binding protein required for initiation of protein translation). Further, molecular docking and molecular dynamics simulation studies suggested that emetine may bind to the cap-binding pocket of eIF4E, in a similar conformation as m7-GTP binds. Additionally, SARS-CoV-2 was shown to exploit ERK/MNK1/eIF4E signalling pathway for its effective replication in the target cells. Collectively our results suggest that further detailed evaluation of emetine as a potential treatment for COVID-19 may be warranted.


Subject(s)
Antiviral Agents , Emetine , Infectious bronchitis virus/drug effects , RNA, Viral/metabolism , SARS-CoV-2/drug effects , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Chick Embryo , Chlorocebus aethiops , Coronavirus Infections/drug therapy , Emetine/pharmacology , Emetine/therapeutic use , Eukaryotic Initiation Factor-4E/metabolism , Protein Binding/drug effects , RNA, Messenger/metabolism , Signal Transduction , Vero Cells
12.
Emerg Microbes Infect ; 10(1): 481-492, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1124537

ABSTRACT

The unprecedented coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a serious threat to global public health. Development of effective therapies against SARS-CoV-2 is urgently needed. Here, we evaluated the antiviral activity of a remdesivir parent nucleotide analog, GS441524, which targets the coronavirus RNA-dependent RNA polymerase enzyme, and a feline coronavirus prodrug, GC376, which targets its main protease, using a mouse-adapted SARS-CoV-2 infected mouse model. Our results showed that GS441524 effectively blocked the proliferation of SARS-CoV-2 in the mouse upper and lower respiratory tracts via combined intranasal (i.n.) and intramuscular (i.m.) treatment. However, the ability of high-dose GC376 (i.m. or i.n. and i.m.) was weaker than GS441524. Notably, low-dose combined application of GS441524 with GC376 could effectively protect mice against SARS-CoV-2 infection via i.n. or i.n. and i.m. treatment. Moreover, we found that the pharmacokinetic properties of GS441524 is better than GC376, and combined application of GC376 and GS441524 had a synergistic effect. Our findings support the further evaluation of the combined application of GC376 and GS441524 in future clinical studies.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Coronavirus RNA-Dependent RNA Polymerase/antagonists & inhibitors , Respiratory System/virology , SARS-CoV-2/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Cell Proliferation/drug effects , Chlorocebus aethiops , Drug Therapy, Combination , Female , Mice , Mice, Inbred BALB C , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Vero Cells
13.
Emerg Microbes Infect ; 9(1): 1744-1747, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-653863

ABSTRACT

The spread of novel coronavirus disease 2019 (COVID-19) infections worldwide has raised concerns about the prevention and control of SARS-CoV-2. Devices that rapidly inactivate viruses can reduce the chance of infection through aerosols and contact transmission. This in vitro study demonstrated that irradiation with a deep ultraviolet light-emitting diode (DUV-LED) of 280 ± 5 nm wavelength rapidly inactivates SARS-CoV-2 obtained from a COVID-19 patient. Development of devices equipped with DUV-LED is expected to prevent virus invasion through the air and after touching contaminated objects.


Subject(s)
Betacoronavirus/radiation effects , Coronavirus Infections/virology , Pneumonia, Viral/virology , Animals , Betacoronavirus/isolation & purification , COVID-19 , Cell Survival , Chlorocebus aethiops , Decontamination , Humans , Pandemics , SARS-CoV-2 , Ultraviolet Rays , Vero Cells , Virus Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL