71 results on '"Bojkova D"'
Search Results
2. Infectivity of deceased COVID-19 patients
- Author
-
Plenzig, Stefanie, Bojkova, D., Held, H., Berger, A., Holz, F., Cinatl, J., Gradhand, E., Kettner, M., Pfeiffer, A., Verhoff, M. A., and Ciesek, S.
- Published
- 2021
- Full Text
- View/download PDF
3. Short Lecture “Natural products against SARS-CoV-2 or how to catch a butterfly?”
- Author
-
Wasilewicz, A, primary, Kirchweger, B, additional, Bojkova, D, additional, Jose Abi Saad, M, additional, Langeder, J, additional, Bütikofer, M, additional, Grienke, U, additional, Cinatl, J, additional, Orts, J, additional, Kirchmair, J, additional, Rabenau, H, additional, and Rollinger, J M, additional
- Published
- 2022
- Full Text
- View/download PDF
4. Novel SARS-CoV-2 variants induce higher toxicity in cardiovascular cells
- Author
-
Wagner, J U G, primary, Bojkova, D, additional, Shumliakivska, M, additional, Aslan, G S, additional, Kandler, J D, additional, Hansen, A, additional, Krishnan, J, additional, Zeiher, A M, additional, Ciesek, S, additional, Cinatl, J, additional, and Dimmeler, S, additional
- Published
- 2021
- Full Text
- View/download PDF
5. COVID-19-related coagulopathy - is transferrin a missing link?
- Author
-
McLaughlin, K.-M., Bechtel, M., Bojkova, D., Münch, C., Ciesek, S., Wass, M.N., Michaelis, M., Cinatl, J., and Publica
- Subjects
ddc:610 - Abstract
SARS-CoV-2 is the causative agent of COVID-19. Severe COVID-19 disease has been associated with disseminated intravascular coagulation and thrombosis, but the mechanisms underlying COVID-19-related coagulopathy remain unknown. The risk of severe COVID-19 disease is higher in males than in females and increases with age. To identify gene products that may contribute to COVID-19-related coagulopathy, we analyzed the expression of genes associated with the Gene Ontology (GO) term ""blood coagulation"" in the Genotype-Tissue Expression (GTEx) database and identified four procoagulants, whose expression is higher in males and increases with age (ADAMTS13, F11, HGFAC, KLKB1), and two anticoagulants, whose expression is higher in females and decreases with age (C1QTNF1, SERPINA5). However, the expression of none of these genes was regulated in a proteomics dataset of SARS-CoV-2-infected cells and none of the proteins have been identified as a binding partner of SARS-CoV-2 proteins. Hence, they may rather generally predispose individuals to thrombosis without directly contributing to COVID-19-related coagulopathy. In contrast, the expression of the procoagulant transferrin (not associated to the GO term ""blood coagulation"") was higher in males, increased with age, and was upregulated upon SARS-CoV-2 infection. Hence, transferrin warrants further examination in ongoing clinic-pathological investigations.
- Published
- 2020
6. Expanding the donor pool-decontamination of HCV RNA positive kidneys with methylene blue
- Author
-
Helfritz, F., primary, Bojkova, D., additional, Westhaus, S., additional, Steinmann, E., additional, Minor, T., additional, Meuleman, P., additional, Paul, A., additional, and Ciesek, S., additional
- Published
- 2018
- Full Text
- View/download PDF
7. SAT-348 - Expanding the donor pool-decontamination of HCV RNA positive kidneys with methylene blue
- Author
-
Helfritz, F., Bojkova, D., Westhaus, S., Steinmann, E., Minor, T., Meuleman, P., Paul, A., and Ciesek, S.
- Published
- 2018
- Full Text
- View/download PDF
8. Study of liquid–liquid extraction of perrhenate with cyclohexanone in different media
- Author
-
Jordanov, N., Pavlova, M., and Bojkova, D.
- Published
- 1976
- Full Text
- View/download PDF
9. Correction: Biochemical characterization of protease activity of Nsp3 from SARS-CoV-2 and its inhibition by nanobodies.
- Author
-
Armstrong LA, Lange SM, Cesare V, Matthews SP, Nirujogi RS, Cole I, Hope A, Cunningham F, Toth R, Mukherjee R, Bojkova D, Gruber F, Gray D, Wyatt PG, Cinatl J, Dikic I, Davies P, and Kulathu Y
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0253364.]., (Copyright: © 2024 Armstrong et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
10. Molecular networking unveils anti-SARS-CoV-2 constituents from traditionally used remedies.
- Author
-
Wasilewicz A, Bojkova D, Beniddir MA, Cinatl J Jr, Rabenau HF, Grienke U, Rollinger JM, and Kirchweger B
- Subjects
- Humans, Caco-2 Cells, Post-Acute COVID-19 Syndrome, Plant Extracts therapeutic use, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, SARS-CoV-2, COVID-19
- Abstract
Ethnopharmacological Relevance: Plants and fungi have a long tradition in ethnopharmacology for the treatment of infectious diseases including viruses. Many of these natural products have also been used to combat SARS-CoV-2 infections or symptoms of the post- and long-COVID form, owing to the scarcity of clinically approved therapeutics., Aim of the Study: The ongoing threat posed by SARS-CoV-2, along with the rapidly evolving new variants, requires the development of new antiviral compounds. The aim of this study was to identify anti-SARS-CoV-2 herbal and fungal extracts used in traditional medicine against acute respiratory infection, inflammation, and related symptoms. Additionally, we sought to characterize their bioactive constituents., Materials and Methods: The antiviral activity and cell cytotoxicity of 179 herbal and fungal extracts were evaluated using two SARS-CoV-2 infection assays in Caco-2 cells. 19 plant extracts with and without anti-SARS-CoV-2 activity underwent detailed dereplication using molecular networking., Results: Extracts from Angelica sinensis (Oliv.) Diels roots, Annona squamosa L. seeds, Azadirachta indica A. Juss. fruits, Buddleja officinalis Maxim. flowers, Burkea africana Hook. bark and Clinopodium menthifolium (Host) Stace aerial parts showed a potent anti SARS-CoV-2 activity (IC
50 < 5 μg/ml) with only moderate cytotoxicity (CC50 > 60 μg/ml, Caco-2). By performing the dereplication with a bioactivity-featured molecular network (MN) on the extract library level, rather than on the level of individual extracts, we could pinpoint compounds characteristic for active extracts. Thus, a straight-forward identification of potential anti-SARS-CoV-2 natural compounds was achieved prior to any fractionation or isolation efforts., Conclusions: A sophisticated hyphenation of empirical knowledge with MS-based bioinformatics and automated compound annotation was applied to decipher the chemical space of the investigated extracts. The correlation with experimentally assessed anti-SARS-CoV-2 activities helped in predicting compound classes and structural elements relevant for the antiviral activities. Consequently, this accelerated the identification of constituents from the investigated mixtures with inhibitory effects against SARS-CoV-2., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
11. Trifluridine for treatment of mpox infection in drug combinations in ophthalmic cell models.
- Author
-
Cinatl J, Bechtel M, Reus P, Ott M, Rothweiler F, Michaelis M, Ciesek S, and Bojkova D
- Subjects
- Humans, Eye, Drug Combinations, Benzamides, Isoindoles, Monkeypox virus, Trifluridine pharmacology, Trifluridine therapeutic use, Mpox (monkeypox)
- Abstract
The Mpox virus can cause severe disease in the susceptible population with dermatologic and systemic manifestations. Furthermore, ophthalmic manifestations of mpox infection are well documented. Topical trifluridine (TFT) eye drops have been used for therapy of ophthalmic mpox infection in patients, however, its efficacy against mpox virus infection in this scenario has not been previously shown. In the present study, we have established ophthalmic cell models suitable for the infection with mpox virus. We show, that TFT is effective against a broad range of mpox isolates in conjunctival epithelial cells and keratocytes. Further, TFT remained effective against a tecovirimat-resistant virus strain. In the context of drug combinations, a nearly additive effect was observed for TFT combinations with brincidofovir and tecovirimat in conjunctival epithelial cells, while a slight antagonism was observed for both combinations in keratocytes. Altogether, our findings demonstrate TFT as a promising drug for treatment of ophthalmic mpox infection able to overcome tecovirimat resistance. However, conflicting results regarding the effect of drug combinations with approved compounds warrant close monitoring of such use in patients., (© 2024 The Authors. Journal of Medical Virology published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
12. Drug repurposing for the treatment of COVID-19: Targeting nafamostat to the lungs by a liposomal delivery system.
- Author
-
Reus P, Guthmann H, Uhlig N, Agbaria M, Issmail L, Eberlein V, Nordling-David MM, Jbara-Agbaria D, Ciesek S, Bojkova D, Cinatl J, Burger-Kentischer A, Rupp S, Zaliani A, Grunwald T, Gribbon P, Kannt A, and Golomb G
- Subjects
- Humans, Mice, Animals, Liposomes, Drug Repositioning, Pandemics, Tissue Distribution, Lung, SARS-CoV-2, COVID-19
- Abstract
Despite tremendous global efforts since the beginning of the COVID-19 pandemic, still only a limited number of prophylactic and therapeutic options are available. Although vaccination is the most effective measure in preventing morbidity and mortality, there is a need for safe and effective post-infection treatment medication. In this study, we explored a pipeline of 21 potential candidates, examined in the Calu-3 cell line for their antiviral efficacy, for drug repurposing. Ralimetinib and nafamostat, clinically used drugs, have emerged as attractive candidates. Due to the inherent limitations of the selected drugs, we formulated targeted liposomes suitable for both systemic and intranasal administration. Non-targeted and targeted nafamostat liposomes (LipNaf) decorated with an Apolipoprotein B peptide (ApoB-P) as a specific lung-targeting ligand were successfully developed. The developed liposomal formulations of nafamostat were found to possess favorable physicochemical properties including nano size (119-147 nm), long-term stability of the normally rapidly degrading compound in aqueous solution, negligible leakage from the liposomes upon storage, and a neutral surface charge with low polydispersity index (PDI). Both nafamostat and ralimetinib liposomes showed good cellular uptake and lack of cytotoxicity, and non-targeted LipNaf demonstrated enhanced accumulation in the lungs following intranasal (IN) administration in non-infected mice. LipNaf retained its anti-SARS-CoV 2 activity in Calu 3 cells with only a modest decrease, exhibiting complete inhibition at concentrations >100 nM. IN, but not intraperitoneal (IP) treatment with targeted LipNaf resulted in a trend to reduced viral load in the lungs of K18-hACE2 mice compared to targeted empty Lip. Nevertheless, upon removal of outlier data, a statistically significant 1.9-fold reduction in viral load was achieved. This observation further highlights the importance of a targeted delivery into the respiratory tract. In summary, we were able to demonstrate a proof-of-concept of drug repurposing by liposomal formulations with anti-SARS-CoV-2 activity. The biodistribution and bioactivity studies with LipNaf suggest an IN or inhalation route of administration for optimal therapeutic efficacy., Competing Interests: Declaration of Competing Interest The authors declare no conflicts of interest., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. Discovery of anti-SARS-CoV-2 secondary metabolites from the heartwood of Pterocarpus santalinus using multi-informative molecular networking.
- Author
-
Wasilewicz A, Zwirchmayr J, Kirchweger B, Bojkova D, Cinatl J Jr, Rabenau HF, Rollinger JM, Beniddir MA, and Grienke U
- Abstract
A pigment-depleted extract from the heartwood of Pterocarpus santalinus L. f. (PS-DE) showed promising anti-SARS-CoV-2 activity with an IC
50 of 29.9 μg/mL in Caco-2-F03 cells. To determine the potential active constituents within the extract prior to isolation, multi-informative molecular network (MN) was applied. Therefore, the extract was separated by high-performance counter-current chromatography (HPCCC) into 11 fractions which were subsequently tested for anti-SARS-CoV-2 activity and analysed by UPLC-tandem mass spectrometry (MS2 ). The resulting MN combines the bioactivity data of the fractions with the MS2 data. The MN analysis led to the targeted isolation of seven compounds including one pterocarpan (7) reported for the first time as constituent of P. santalinus and four so far undescribed natural products (NPs) that belong to the compound classes of arylpropanes (9) , isoflavanones (10) coumestans (16) and 3-arylcoumarins (17) , respectively. In total, 15 constituents from the heartwood of P. santalinus and one synthetic isoflavonoid that is structurally related to the natural metabolites were tested for anti-SARS-CoV-2 activity. Thereby, the two pterocarpans (-)-homopterocarpin (5) and (-)-medicarpin (2) , the stilbene (E) -pterostilbene (1) and the isoflavonoid 7-O-methylgenistein (11) showed a distinct antiviral activity with IC50 values of 17.2, 33.4, 34.7, and 37.9 µM, respectively, and no cytotoxic effects against Caco-2-F03 cells (CC50 > 100 µM). In addition, a structure-activity relationship (SAR) was proposed indicating structural requirements of pterocarpans for anti-SARS-CoV-2 activity. The herein presented results support the implementation of multi-informative molecular networks as powerful tool for dereplication and targeted isolation of bioactive NPs., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Wasilewicz, Zwirchmayr, Kirchweger, Bojkova, Cinatl, Rabenau, Rollinger, Beniddir and Grienke.)- Published
- 2023
- Full Text
- View/download PDF
14. Silver N-heterocyclic carbene complexes are potent uncompetitive inhibitors of the papain-like protease with antiviral activity against SARS-CoV-2.
- Author
-
Gil-Moles M, O'Beirne C, Esarev IV, Lippmann P, Tacke M, Cinatl J Jr, Bojkova D, and Ott I
- Abstract
The ongoing SARS-CoV-2 pandemic has caused a high demand for novel innovative antiviral drug candidates. Despite promising results, metal complexes have been relatively unexplored as antiviral agents in general and in particular against SARS-CoV-2. Here we report on silver NHC complexes with chloride or iodide counter ligands that are potent inhibitors of the SARS-CoV-2 papain-like protease (PL
pro ) but inactive against 3C-like protease (3CLpro ) as another SARS-CoV-2 protease. Mechanistic studies on a selected complex confirmed zinc removal from a zinc binding domain of PLpro as relevant factor of their activity. In addition, enzyme kinetic experiments revealed that the complex is an uncompetitive inhibitor and with this rare type of inhibition it offers great pharmacological advantages in terms selectivity. The silver NHC complexes with iodide ligands showed very low or absent host cell toxicity and triggered strong effects on viral replication in cells infected with SARS-CoV-2, making them promising future antiviral drug candidates., Competing Interests: There is no conflict of interest to declare., (This journal is © The Royal Society of Chemistry.)- Published
- 2023
- Full Text
- View/download PDF
15. SARS-CoV-2 Variants Show Different Host Cell Proteome Profiles With Delayed Immune Response Activation in Omicron-Infected Cells.
- Author
-
Metzler M, Tharyan RG, Klann K, Grikscheit K, Bojkova D, Cinatl J, Tascher G, Ciesek S, and Münch C
- Subjects
- Humans, Proteome, Pandemics, Antiviral Agents, Antibodies, Neutralizing, SARS-CoV-2, COVID-19
- Abstract
The ancestral SARS-CoV-2 strain that initiated the Covid-19 pandemic at the end of 2019 has rapidly mutated into multiple variants of concern with variable pathogenicity and increasing immune escape strategies. However, differences in host cellular antiviral responses upon infection with SARS-CoV-2 variants remain elusive. Leveraging whole-cell proteomics, we determined host signaling pathways that are differentially modulated upon infection with the clinical isolates of the ancestral SARS-CoV-2 B.1 and the variants of concern Delta and Omicron BA.1. Our findings illustrate alterations in the global host proteome landscape upon infection with SARS-CoV-2 variants and the resulting host immune responses. Additionally, viral proteome kinetics reveal declining levels of viral protein expression during Omicron BA.1 infection when compared to ancestral B.1 and Delta variants, consistent with its reduced replication rates. Moreover, molecular assays reveal deferral activation of specific host antiviral signaling upon Omicron BA.1 and BA.2 infections. Our study provides an overview of host proteome profile of multiple SARS-CoV-2 variants and brings forth a better understanding of the instigation of key immune signaling pathways causative for the differential pathogenicity of SARS-CoV-2 variants., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Identification of Z-Tyr-Ala-CHN 2 , a Cathepsin L Inhibitor with Broad-Spectrum Cell-Specific Activity against Coronaviruses, including SARS-CoV-2.
- Author
-
Doijen J, Temmerman K, Van den Eynde C, Diels A, Van den Broeck N, Van Gool M, Heo I, Jaensch S, Zwaagstra M, Diosa Toro M, Chiu W, De Jonghe S, Leyssen P, Bojkova D, Ciesek S, Cinatl J, Verschueren L, Buyck C, Van Kuppeveld F, Neyts J, Van Loock M, and Van Damme E
- Abstract
The ongoing COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is partly under control by vaccination. However, highly potent and safe antiviral drugs for SARS-CoV-2 are still needed to avoid development of severe COVID-19. We report the discovery of a small molecule, Z-Tyr-Ala-CHN
2 , which was identified in a cell-based antiviral screen. The molecule exerts sub-micromolar antiviral activity against SARS-CoV-2, SARS-CoV-1, and human coronavirus 229E. Time-of-addition studies reveal that Z-Tyr-Ala-CHN2 acts at the early phase of the infection cycle, which is in line with the observation that the molecule inhibits cathepsin L. This results in antiviral activity against SARS-CoV-2 in VeroE6, A549-hACE2, and HeLa-hACE2 cells, but not in Caco-2 cells or primary human nasal epithelial cells since the latter two cell types also permit entry via transmembrane protease serine subtype 2 (TMPRSS2). Given their cell-specific activity, cathepsin L inhibitors still need to prove their value in the clinic; nevertheless, the activity profile of Z-Tyr-Ala-CHN2 makes it an interesting tool compound for studying the biology of coronavirus entry and replication.- Published
- 2023
- Full Text
- View/download PDF
17. Repurposing of the antibiotic nitroxoline for the treatment of mpox.
- Author
-
Bojkova D, Zöller N, Tietgen M, Steinhorst K, Bechtel M, Rothenburger T, Kandler JD, Schneider J, Corman VM, Ciesek S, Rabenau HF, Wass MN, Kippenberger S, Göttig S, Michaelis M, and Cinatl J Jr
- Subjects
- Humans, Anti-Bacterial Agents pharmacology, Antiviral Agents pharmacology, Cidofovir, Drug Repositioning, Mpox (monkeypox) drug therapy, Nitroquinolines pharmacology
- Abstract
The antiviral drugs tecovirimat, brincidofovir, and cidofovir are considered for mpox (monkeypox) treatment despite a lack of clinical evidence. Moreover, their use is affected by toxic side-effects (brincidofovir, cidofovir), limited availability (tecovirimat), and potentially by resistance formation. Hence, additional, readily available drugs are needed. Here, therapeutic concentrations of nitroxoline, a hydroxyquinoline antibiotic with a favourable safety profile in humans, inhibited the replication of 12 mpox virus isolates from the current outbreak in primary cultures of human keratinocytes and fibroblasts and a skin explant model by interference with host cell signalling. Tecovirimat, but not nitroxoline, treatment resulted in rapid resistance development. Nitroxoline remained effective against the tecovirimat-resistant strain and increased the anti-mpox virus activity of tecovirimat and brincidofovir. Moreover, nitroxoline inhibited bacterial and viral pathogens that are often co-transmitted with mpox. In conclusion, nitroxoline is a repurposing candidate for the treatment of mpox due to both antiviral and antimicrobial activity., (© 2023 The Authors. Journal of Medical Virology published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
18. Omicron-induced interferon signaling prevents influenza A H1N1 and H5N1 virus infection.
- Author
-
Bojkova D, Bechtel M, Rothenburger T, Kandler JD, Hayes L, Olmer R, Martin U, Jonigk D, Ciesek S, Wass MN, Michaelis M, and Cinatl J Jr
- Subjects
- Humans, SARS-CoV-2, Interferons, Antiviral Agents, Influenza A Virus, H5N1 Subtype, Influenza, Human, Influenza A Virus, H1N1 Subtype, COVID-19, Influenza A virus, Janus Kinase Inhibitors
- Abstract
Recent findings in permanent cell lines suggested that SARS-CoV-2 Omicron BA.1 induces a stronger interferon response than Delta. Here, we show that BA.1 and BA.5 but not Delta induce an antiviral state in air-liquid interface cultures of primary human bronchial epithelial cells and primary human monocytes. Both Omicron subvariants caused the production of biologically active types I (α/β) and III (λ) interferons and protected cells from super-infection with influenza A viruses. Notably, abortive Omicron infection of monocytes was sufficient to protect monocytes from influenza A virus infection. Interestingly, while influenza-like illnesses surged during the Delta wave in England, their spread rapidly declined upon the emergence of Omicron. Mechanistically, Omicron-induced interferon signaling was mediated via double-stranded RNA recognition by MDA5, as MDA5 knockout prevented it. The JAK/STAT inhibitor baricitinib inhibited the Omicron-mediated antiviral response, suggesting it is caused by MDA5-mediated interferon production, which activates interferon receptors that then trigger JAK/STAT signaling. In conclusion, our study (1) demonstrates that only Omicron but not Delta induces a substantial interferon response in physiologically relevant models, (2) shows that Omicron infection protects cells from influenza A virus super-infection, and (3) indicates that BA.1 and BA.5 induce comparable antiviral states., (© 2023 The Authors. Journal of Medical Virology published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
19. A Monoclonal Human Alveolar Epithelial Cell Line ("Arlo") with Pronounced Barrier Function for Studying Drug Permeability and Viral Infections.
- Author
-
Carius P, Jungmann A, Bechtel M, Grißmer A, Boese A, Gasparoni G, Salhab A, Seipelt R, Urbschat K, Richter C, Meier C, Bojkova D, Cinatl J, Walter J, Schneider-Daum N, and Lehr CM
- Subjects
- Animals, Humans, SARS-CoV-2, Cell Line, Permeability, Alveolar Epithelial Cells, COVID-19 metabolism
- Abstract
In the development of orally inhaled drug products preclinical animal models regularly fail to predict pharmacological as well as toxicological responses in humans. Models based on human cells and tissues are potential alternatives to animal experimentation allowing for the isolation of essential processes of human biology and making them accessible in vitro. Here, the generation of a novel monoclonal cell line "Arlo," derived from the polyclonal human alveolar epithelium lentivirus immortalized cell line hAELVi via single-cell printing, and its characterization as a model for the human alveolar epithelium as well as a building block for future complex in vitro models is described. "Arlo" is systematically compared in vitro to primary human alveolar epithelial cells (hAEpCs) as well as to the polyclonal hAELVi cell line. "Arlo" cells show enhanced barrier properties with high transepithelial electrical resistance (TEER) of ≈3000 Ω cm
2 and a potential difference (PD) of ≈30 mV under air-liquid interface (ALI) conditions, that can be modulated. The cells grow in a polarized monolayer and express genes relevant to barrier integrity as well as homeostasis as is observed in hAEpCs. Successful productive infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a proof-of-principle study offers an additional, attractive application of "Arlo" beyond biopharmaceutical experimentation., (© 2023 The Authors. Advanced Science published by Wiley-VCH GmbH.)- Published
- 2023
- Full Text
- View/download PDF
20. Identification of Natural Products Inhibiting SARS-CoV-2 by Targeting Viral Proteases: A Combined in Silico and in Vitro Approach.
- Author
-
Wasilewicz A, Kirchweger B, Bojkova D, Abi Saad MJ, Langeder J, Bütikofer M, Adelsberger S, Grienke U, Cinatl J Jr, Petermann O, Scapozza L, Orts J, Kirchmair J, Rabenau HF, and Rollinger JM
- Subjects
- Humans, Viral Proteases, SARS-CoV-2, Peptide Hydrolases, Antiviral Agents, Molecular Docking Simulation, Protease Inhibitors, COVID-19, Biological Products
- Abstract
In this study, an integrated in silico-in vitro approach was employed to discover natural products (NPs) active against SARS-CoV-2. The two SARS-CoV-2 viral proteases, i.e., main protease (M
pro ) and papain-like protease (PLpro ), were selected as targets for the in silico study. Virtual hits were obtained by docking more than 140,000 NPs and NP derivatives available in-house and from commercial sources, and 38 virtual hits were experimentally validated in vitro using two enzyme-based assays. Five inhibited the enzyme activity of SARS-CoV-2 Mpro by more than 60% at a concentration of 20 μM, and four of them with high potency (IC50 < 10 μM). These hit compounds were further evaluated for their antiviral activity against SARS-CoV-2 in Calu-3 cells. The results from the cell-based assay revealed three mulberry Diels-Alder-type adducts (MDAAs) from Morus alba with pronounced anti-SARS-CoV-2 activities. Sanggenons C ( 12 ), O ( 13 ), and G ( 15 ) showed IC50 values of 4.6, 8.0, and 7.6 μM and selectivity index values of 5.1, 3.1 and 6.5, respectively. The docking poses of MDAAs in SARS-CoV-2 Mpro proposed a butterfly-shaped binding conformation, which was supported by the results of saturation transfer difference NMR experiments and competitive1 H relaxation dispersion NMR spectroscopy.- Published
- 2023
- Full Text
- View/download PDF
21. Identification of novel antiviral drug candidates using an optimized SARS-CoV-2 phenotypic screening platform.
- Author
-
Bojkova D, Reus P, Panosch L, Bechtel M, Rothenburger T, Kandler JD, Pfeiffer A, Wagner JUG, Shumliakivska M, Dimmeler S, Olmer R, Martin U, Vondran FWR, Toptan T, Rothweiler F, Zehner R, Rabenau HF, Osman KL, Pullan ST, Carroll MW, Stack R, Ciesek S, Wass MN, Michaelis M, and Cinatl J Jr
- Abstract
Reliable, easy-to-handle phenotypic screening platforms are needed for the identification of anti-SARS-CoV-2 compounds. Here, we present caspase 3/7 activity as a readout for monitoring the replication of SARS-CoV-2 isolates from different variants, including a remdesivir-resistant strain, and of other coronaviruses in numerous cell culture models, independently of cytopathogenic effect formation. Compared to other models, the Caco-2 subline Caco-2-F03 displayed superior performance. It possesses a stable SARS-CoV-2 susceptibility phenotype and does not produce false-positive hits due to drug-induced phospholipidosis. A proof-of-concept screen of 1,796 kinase inhibitors identified known and novel antiviral drug candidates including inhibitors of phosphoglycerate dehydrogenase (PHGDH), CDC like kinase 1 (CLK-1), and colony stimulating factor 1 receptor (CSF1R). The activity of the PHGDH inhibitor NCT-503 was further increased in combination with the hexokinase II (HK2) inhibitor 2-deoxy-D-glucose, which is in clinical development for COVID-19. In conclusion, caspase 3/7 activity detection in SARS-CoV-2-infected Caco-2-F03 cells provides a simple phenotypic high-throughput screening platform for SARS-CoV-2 drug candidates that reduces false-positive hits., Competing Interests: The authors declare no competing interests., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
22. Drug Sensitivity of Currently Circulating Mpox Viruses.
- Author
-
Bojkova D, Bechtel M, Rothenburger T, Steinhorst K, Zöller N, Kippenberger S, Schneider J, Corman VM, Uri H, Wass MN, Knecht G, Khaykin P, Wolf T, Ciesek S, Rabenau HF, Michaelis M, and Cinatl J Jr
- Subjects
- Humans, Drug Resistance, Viral physiology, Mpox (monkeypox) drug therapy, Mpox (monkeypox) physiopathology, Mpox (monkeypox) virology, Monkeypox virus drug effects, Monkeypox virus physiology, Antiviral Agents pharmacology
- Published
- 2023
- Full Text
- View/download PDF
23. Impact of the COVID-19 pandemic on the circulation of other pathogens in England.
- Author
-
Hayes LJ, Uri H, Bojkova D, Cinatl J Jr, Wass MN, and Michaelis M
- Subjects
- Humans, Pandemics, England epidemiology, COVID-19 epidemiology
- Published
- 2023
- Full Text
- View/download PDF
24. Synergism of interferon-beta with antiviral drugs against SARS-CoV-2 variants.
- Author
-
Bojkova D, Stack R, Rothenburger T, Kandler JD, Ciesek S, Wass MN, Michaelis M, and Cinatl J Jr
- Subjects
- Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Humans, Interferon-beta therapeutic use, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
Competing Interests: Declaration of Competing Interests The authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
25. USP22 controls type III interferon signaling and SARS-CoV-2 infection through activation of STING.
- Author
-
Karlowitz R, Stanifer ML, Roedig J, Andrieux G, Bojkova D, Bechtel M, Smith S, Kowald L, Schubert R, Boerries M, Cinatl J Jr, Boulant S, and van Wijk SJL
- Subjects
- Antiviral Agents pharmacology, Humans, Interferons metabolism, Pandemics, SARS-CoV-2, Interferon Lambda, COVID-19, Interferon Type I genetics, Membrane Proteins metabolism, Ubiquitin Thiolesterase metabolism
- Abstract
Pattern recognition receptors (PRRs) and interferons (IFNs) serve as essential antiviral defense against SARS-CoV-2, the causative agent of the COVID-19 pandemic. Type III IFNs (IFN-λ) exhibit cell-type specific and long-lasting functions in auto-inflammation, tumorigenesis, and antiviral defense. Here, we identify the deubiquitinating enzyme USP22 as central regulator of basal IFN-λ secretion and SARS-CoV-2 infections in human intestinal epithelial cells (hIECs). USP22-deficient hIECs strongly upregulate genes involved in IFN signaling and viral defense, including numerous IFN-stimulated genes (ISGs), with increased secretion of IFN-λ and enhanced STAT1 signaling, even in the absence of exogenous IFNs or viral infection. Interestingly, USP22 controls basal and 2'3'-cGAMP-induced STING activation and loss of STING reversed STAT activation and ISG and IFN-λ expression. Intriguingly, USP22-deficient hIECs are protected against SARS-CoV-2 infection, viral replication, and the formation of de novo infectious particles, in a STING-dependent manner. These findings reveal USP22 as central host regulator of STING and type III IFN signaling, with important implications for SARS-CoV-2 infection and antiviral defense., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
26. Development and optimization of a high-throughput screening assay for in vitro anti-SARS-CoV-2 activity: Evaluation of 5676 Phase 1 Passed Structures.
- Author
-
Chiu W, Verschueren L, Van den Eynde C, Buyck C, De Meyer S, Jochmans D, Bojkova D, Ciesek S, Cinatl J, De Jonghe S, Leyssen P, Neyts J, Van Loock M, and Van Damme E
- Subjects
- Animals, Humans, Antiviral Agents pharmacology, Caco-2 Cells, Chlorocebus aethiops, High-Throughput Screening Assays, Pandemics, COVID-19 Drug Treatment, SARS-CoV-2
- Abstract
Although vaccines are currently used to control the coronavirus disease 2019 (COVID-19) pandemic, treatment options are urgently needed for those who cannot be vaccinated and for future outbreaks involving new severe acute respiratory syndrome coronavirus virus 2 (SARS-CoV-2) strains or coronaviruses not covered by current vaccines. Thus far, few existing antivirals are known to be effective against SARS-CoV-2 and clinically successful against COVID-19. As part of an immediate response to the COVID-19 pandemic, a high-throughput, high content imaging-based SARS-CoV-2 infection assay was developed in VeroE6 African green monkey kidney epithelial cells expressing a stable enhanced green fluorescent protein (VeroE6-eGFP cells) and was used to screen a library of 5676 compounds that passed Phase 1 clinical trials. Eight drugs (nelfinavir, RG-12915, itraconazole, chloroquine, hydroxychloroquine, sematilide, remdesivir, and doxorubicin) were identified as inhibitors of in vitro anti-SARS-CoV-2 activity in VeroE6-eGFP and/or Caco-2 cell lines. However, apart from remdesivir, toxicity and pharmacokinetic data did not support further clinical development of these compounds for COVID-19 treatment., (© 2022 Janssen Pharmaceutica NV. Journal of Medical Virology published by Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
27. SARS-CoV-2 Omicron variant virus isolates are highly sensitive to interferon treatment.
- Author
-
Bojkova D, Rothenburger T, Ciesek S, Wass MN, Michaelis M, and Cinatl J Jr
- Published
- 2022
- Full Text
- View/download PDF
28. SARS-CoV-2 screening strategies for returning international travellers: Evaluation of a rapid antigen test approach.
- Author
-
Layer E, Hoehl S, Widera M, Bojkova D, Westphal T, Gottschalk R, Boeddinghaus B, Schork J, Ciesek S, and Goetsch U
- Subjects
- Humans, Quarantine, Sensitivity and Specificity, Travel, COVID-19 diagnosis, SARS-CoV-2
- Abstract
Background: International travel poses the risk of importing SARS-CoV-2 infections and introducing new viral variants into the country of destination. Established measures include mandatory quarantine with the opportunity to abbreviate it with a negative rapid antigen test (RAT)., Methods: A total of 1,488 returnees were tested for SARS-CoV-2 with both PCR and RAT no earlier than 5 days after arrival. We assessed the sensitivity and specificity of the RAT. Positive samples were evaluated for infectivity in vitro in a cell culture outgrowth assay. We tracked if participants who tested negative were reported positive within 2 weeks of the initial test., Results: Potential infectiousness was determined based on symptom onset analysis, resulting in a sensitivity of the antigen test of 89% in terms of infectivity. The specificity was 100%. All positive outgrowth assays were preceded by a positive RAT, indicating that all participants with proven in vitro infectivity were correctly identified. None of the negative participants tested positive during the follow-up., Conclusions: RAT no earlier than the 5th day after arrival was a reliable method for detecting infectious travellers and can be recommended as an appropriate method for managing SARS-CoV-2 travel restrictions. Compliance to the regulations and a high standard of test quality must be ensured., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Reduced interferon antagonism but similar drug sensitivity in Omicron variant compared to Delta variant of SARS-CoV-2 isolates.
- Author
-
Bojkova D, Widera M, Ciesek S, Wass MN, Michaelis M, and Cinatl J Jr
- Subjects
- Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Humans, Interferons, SARS-CoV-2, COVID-19 Drug Treatment
- Published
- 2022
- Full Text
- View/download PDF
30. Ibuprofen, Flurbiprofen, Etoricoxib or Paracetamol Do Not Influence ACE2 Expression and Activity In Vitro or in Mice and Do Not Exacerbate In-Vitro SARS-CoV-2 Infection.
- Author
-
de Bruin N, Schneider AK, Reus P, Talmon S, Ciesek S, Bojkova D, Cinatl J, Lodhi I, Charlesworth B, Sinclair S, Pennick G, Laughey WF, Gribbon P, Kannt A, and Schiffmann S
- Subjects
- Acetaminophen pharmacology, Animals, COVID-19 metabolism, COVID-19 pathology, Caco-2 Cells, Disease Progression, Enzyme Activation drug effects, Etoricoxib pharmacology, Flurbiprofen pharmacology, Gene Expression Regulation, Enzymologic drug effects, Humans, Ibuprofen pharmacology, Male, Mice, Mice, Inbred C57BL, SARS-CoV-2 drug effects, SARS-CoV-2 physiology, Virus Internalization drug effects, Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 metabolism, Anti-Inflammatory Agents, Non-Steroidal pharmacology, COVID-19 genetics
- Abstract
SARS-CoV-2 uses the human cell surface protein angiotensin converting enzyme 2 (ACE2) as the receptor by which it gains access into lung and other tissue. Early in the pandemic, there was speculation that a number of commonly used medications-including ibuprofen and other non-steroidal anti-inflammatory drugs (NSAIDs)-have the potential to upregulate ACE2, thereby possibly facilitating viral entry and increasing the severity of COVID-19. We investigated the influence of the NSAIDS with a range of cyclooxygenase (COX)1 and COX2 selectivity (ibuprofen, flurbiprofen, etoricoxib) and paracetamol on the level of ACE2 mRNA/protein expression and activity as well as their influence on SARS-CoV-2 infection levels in a Caco-2 cell model. We also analysed the ACE2 mRNA/protein levels and activity in lung, heart and aorta in ibuprofen treated mice. The drugs had no effect on ACE2 mRNA/protein expression and activity in the Caco-2 cell model. There was no up-regulation of ACE2 mRNA/protein expression and activity in lung, heart and aorta tissue in ibuprofen-treated mice in comparison to untreated mice. Viral load was significantly reduced by both flurbiprofen and ibuprofen at high concentrations. Ibuprofen, flurbiprofen, etoricoxib and paracetamol demonstrated no effects on ACE2 expression or activity in vitro or in vivo. Higher concentrations of ibuprofen and flurbiprofen reduced SARS-CoV-2 replication in vitro.
- Published
- 2022
- Full Text
- View/download PDF
31. Metallodrug Profiling against SARS-CoV-2 Target Proteins Identifies Highly Potent Inhibitors of the S/ACE2 interaction and the Papain-like Protease PL pro .
- Author
-
Gil-Moles M, Türck S, Basu U, Pettenuzzo A, Bhattacharya S, Rajan A, Ma X, Büssing R, Wölker J, Burmeister H, Hoffmeister H, Schneeberg P, Prause A, Lippmann P, Kusi-Nimarko J, Hassell-Hart S, McGown A, Guest D, Lin Y, Notaro A, Vinck R, Karges J, Cariou K, Peng K, Qin X, Wang X, Skiba J, Szczupak Ł, Kowalski K, Schatzschneider U, Hemmert C, Gornitzka H, Milaeva ER, Nazarov AA, Gasser G, Spencer J, Ronconi L, Kortz U, Cinatl J, Bojkova D, and Ott I
- Subjects
- Angiotensin-Converting Enzyme 2, Antiviral Agents pharmacology, Coronavirus Papain-Like Proteases antagonists & inhibitors, SARS-CoV-2 drug effects, Spike Glycoprotein, Coronavirus antagonists & inhibitors
- Abstract
The global spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has called for an urgent need for dedicated antiviral therapeutics. Metal complexes are commonly underrepresented in compound libraries that are used for screening in drug discovery campaigns, however, there is growing evidence for their role in medicinal chemistry. Based on previous results, we have selected more than 100 structurally diverse metal complexes for profiling as inhibitors of two relevant SARS-CoV-2 replication mechanisms, namely the interaction of the spike (S) protein with the ACE2 receptor and the papain-like protease PL
pro . In addition to many well-established types of mononuclear experimental metallodrugs, the pool of compounds tested was extended to approved metal-based therapeutics such as silver sulfadiazine and thiomersal, as well as polyoxometalates (POMs). Among the mononuclear metal complexes, only a small number of active inhibitors of the S/ACE2 interaction was identified, with titanocene dichloride as the only strong inhibitor. However, among the gold and silver containing complexes many turned out to be very potent inhibitors of PLpro activity. Highly promising activity against both targets was noted for many POMs. Selected complexes were evaluated in antiviral SARS-CoV-2 assays confirming activity for gold complexes with N-heterocyclic carbene (NHC) or dithiocarbamato ligands, a silver NHC complex, titanocene dichloride as well as a POM compound. These studies might provide starting points for the design of metal-based SARS-CoV-2 antiviral agents., (© 2021 The Authors. Chemistry - A European Journal published by Wiley-VCH GmbH.)- Published
- 2021
- Full Text
- View/download PDF
32. The SARS-CoV-2 main protease M pro causes microvascular brain pathology by cleaving NEMO in brain endothelial cells.
- Author
-
Wenzel J, Lampe J, Müller-Fielitz H, Schuster R, Zille M, Müller K, Krohn M, Körbelin J, Zhang L, Özorhan Ü, Neve V, Wagner JUG, Bojkova D, Shumliakivska M, Jiang Y, Fähnrich A, Ott F, Sencio V, Robil C, Pfefferle S, Sauve F, Coêlho CFF, Franz J, Spiecker F, Lembrich B, Binder S, Feller N, König P, Busch H, Collin L, Villaseñor R, Jöhren O, Altmeppen HC, Pasparakis M, Dimmeler S, Cinatl J, Püschel K, Zelic M, Ofengeim D, Stadelmann C, Trottein F, Nogueiras R, Hilgenfeld R, Glatzel M, Prevot V, and Schwaninger M
- Subjects
- Animals, Blood-Brain Barrier pathology, Brain pathology, Chlorocebus aethiops, Coronavirus 3C Proteases genetics, Cricetinae, Female, Humans, Intracellular Signaling Peptides and Proteins genetics, Male, Mesocricetus, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Microvessels pathology, SARS-CoV-2 genetics, Vero Cells, Blood-Brain Barrier metabolism, Brain metabolism, Coronavirus 3C Proteases metabolism, Intracellular Signaling Peptides and Proteins metabolism, Microvessels metabolism, SARS-CoV-2 metabolism
- Abstract
Coronavirus disease 2019 (COVID-19) can damage cerebral small vessels and cause neurological symptoms. Here we describe structural changes in cerebral small vessels of patients with COVID-19 and elucidate potential mechanisms underlying the vascular pathology. In brains of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected individuals and animal models, we found an increased number of empty basement membrane tubes, so-called string vessels representing remnants of lost capillaries. We obtained evidence that brain endothelial cells are infected and that the main protease of SARS-CoV-2 (M
pro ) cleaves NEMO, the essential modulator of nuclear factor-κB. By ablating NEMO, Mpro induces the death of human brain endothelial cells and the occurrence of string vessels in mice. Deletion of receptor-interacting protein kinase (RIPK) 3, a mediator of regulated cell death, blocks the vessel rarefaction and disruption of the blood-brain barrier due to NEMO ablation. Importantly, a pharmacological inhibitor of RIPK signaling prevented the Mpro -induced microvascular pathology. Our data suggest RIPK as a potential therapeutic target to treat the neuropathology of COVID-19., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
33. Angiotensin II receptor blocker intake associates with reduced markers of inflammatory activation and decreased mortality in patients with cardiovascular comorbidities and COVID-19 disease.
- Author
-
Cremer S, Pilgram L, Berkowitsch A, Stecher M, Rieg S, Shumliakivska M, Bojkova D, Wagner JUG, Aslan GS, Spinner C, Luxán G, Hanses F, Dolff S, Piepel C, Ruppert C, Guenther A, Rüthrich MM, Vehreschild JJ, Wille K, Haselberger M, Heuzeroth H, Hansen A, Eschenhagen T, Cinatl J, Ciesek S, Dimmeler S, Borgmann S, and Zeiher A
- Subjects
- Adult, Aged, Aged, 80 and over, Angiotensin-Converting Enzyme Inhibitors administration & dosage, Biomarkers blood, Comorbidity, Disease-Free Survival, Female, Humans, Inflammation blood, Inflammation drug therapy, Inflammation mortality, Male, Middle Aged, Severity of Illness Index, Survival Rate, Angiotensin Receptor Antagonists administration & dosage, COVID-19 blood, COVID-19 mortality, Hypertension blood, Hypertension drug therapy, Hypertension mortality, Registries, SARS-CoV-2 metabolism, COVID-19 Drug Treatment
- Abstract
Aims: Patients with cardiovascular comorbidities have a significantly increased risk for a critical course of COVID-19. As the SARS-CoV2 virus enters cells via the angiotensin-converting enzyme receptor II (ACE2), drugs which interact with the renin angiotensin aldosterone system (RAAS) were suspected to influence disease severity., Methods and Results: We analyzed 1946 consecutive patients with cardiovascular comorbidities or hypertension enrolled in one of the largest European COVID-19 registries, the Lean European Open Survey on SARS-CoV-2 (LEOSS) registry. Here, we show that angiotensin II receptor blocker intake is associated with decreased mortality in patients with COVID-19 [OR 0.75 (95% CI 0,59-0.96; p = 0.013)]. This effect was mainly driven by patients, who presented in an early phase of COVID-19 at baseline [OR 0,64 (95% CI 0,43-0,96; p = 0.029)]. Kaplan-Meier analysis revealed a significantly lower incidence of death in patients on an angiotensin receptor blocker (ARB) (n = 33/318;10,4%) compared to patients using an angiotensin-converting enzyme inhibitor (ACEi) (n = 60/348;17,2%) or patients who received neither an ACE-inhibitor nor an ARB at baseline in the uncomplicated phase (n = 90/466; 19,3%; p<0.034). Patients taking an ARB were significantly less frequently reaching the mortality predicting threshold for leukocytes (p<0.001), neutrophils (p = 0.002) and the inflammatory markers CRP (p = 0.021), procalcitonin (p = 0.001) and IL-6 (p = 0.049). ACE2 expression levels in human lung samples were not altered in patients taking RAAS modulators., Conclusion: These data suggest a beneficial effect of ARBs on disease severity in patients with cardiovascular comorbidities and COVID-19, which is linked to dampened systemic inflammatory activity., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
34. Targeting the Pentose Phosphate Pathway for SARS-CoV-2 Therapy.
- Author
-
Bojkova D, Costa R, Reus P, Bechtel M, Jaboreck MC, Olmer R, Martin U, Ciesek S, Michaelis M, and Cinatl J Jr
- Abstract
SARS-CoV-2 is causing the coronavirus disease 2019 (COVID-19) pandemic, for which effective pharmacological therapies are needed. SARS-CoV-2 induces a shift of the host cell metabolism towards glycolysis, and the glycolysis inhibitor 2-deoxy-d-glucose (2DG), which interferes with SARS-CoV-2 infection, is under development for the treatment of COVID-19 patients. The glycolytic pathway generates intermediates that supply the non-oxidative branch of the pentose phosphate pathway (PPP). In this study, the analysis of proteomics data indicated increased transketolase (TKT) levels in SARS-CoV-2-infected cells, suggesting that a role is played by the non-oxidative PPP. In agreement, the TKT inhibitor benfooxythiamine (BOT) inhibited SARS-CoV-2 replication and increased the anti-SARS-CoV-2 activity of 2DG. In conclusion, SARS-CoV-2 infection is associated with changes in the regulation of the PPP. The TKT inhibitor BOT inhibited SARS-CoV-2 replication and increased the activity of the glycolysis inhibitor 2DG. Notably, metabolic drugs like BOT and 2DG may also interfere with COVID-19-associated immunopathology by modifying the metabolism of immune cells in addition to inhibiting SARS-CoV-2 replication. Hence, they may improve COVID-19 therapy outcomes by exerting antiviral and immunomodulatory effects.
- Published
- 2021
- Full Text
- View/download PDF
35. Differences between intrinsic and acquired nucleoside analogue resistance in acute myeloid leukaemia cells.
- Author
-
Rothenburger T, Thomas D, Schreiber Y, Wratil PR, Pflantz T, Knecht K, Digianantonio K, Temple J, Schneider C, Baldauf HM, McLaughlin KM, Rothweiler F, Bilen B, Farmand S, Bojkova D, Costa R, Ferreirós N, Geisslinger G, Oellerich T, Xiong Y, Keppler OT, Wass MN, Michaelis M, and Cinatl J Jr
- Subjects
- Cell Line, Tumor, Drug Resistance, Neoplasm, Humans, Leukemia, Myeloid, Acute drug therapy, Nucleosides pharmacology
- Abstract
Background: SAMHD1 mediates resistance to anti-cancer nucleoside analogues, including cytarabine, decitabine, and nelarabine that are commonly used for the treatment of leukaemia, through cleavage of their triphosphorylated forms. Hence, SAMHD1 inhibitors are promising candidates for the sensitisation of leukaemia cells to nucleoside analogue-based therapy. Here, we investigated the effects of the cytosine analogue CNDAC, which has been proposed to be a SAMHD1 inhibitor, in the context of SAMHD1., Methods: CNDAC was tested in 13 acute myeloid leukaemia (AML) cell lines, in 26 acute lymphoblastic leukaemia (ALL) cell lines, ten AML sublines adapted to various antileukaemic drugs, 24 single cell-derived clonal AML sublines, and primary leukaemic blasts from 24 AML patients. Moreover, 24 CNDAC-resistant sublines of the AML cell lines HL-60 and PL-21 were established. The SAMHD1 gene was disrupted using CRISPR/Cas9 and SAMHD1 depleted using RNAi, and the viral Vpx protein. Forced DCK expression was achieved by lentiviral transduction. SAMHD1 promoter methylation was determined by PCR after treatment of genomic DNA with the methylation-sensitive HpaII endonuclease. Nucleoside (analogue) triphosphate levels were determined by LC-MS/MS. CNDAC interaction with SAMHD1 was analysed by an enzymatic assay and by crystallisation., Results: Although the cytosine analogue CNDAC was anticipated to inhibit SAMHD1, SAMHD1 mediated intrinsic CNDAC resistance in leukaemia cells. Accordingly, SAMHD1 depletion increased CNDAC triphosphate (CNDAC-TP) levels and CNDAC toxicity. Enzymatic assays and crystallisation studies confirmed CNDAC-TP to be a SAMHD1 substrate. In 24 CNDAC-adapted acute myeloid leukaemia (AML) sublines, resistance was driven by DCK (catalyses initial nucleoside phosphorylation) loss. CNDAC-adapted sublines displayed cross-resistance only to other DCK substrates (e.g. cytarabine, decitabine). Cell lines adapted to drugs not affected by DCK or SAMHD1 remained CNDAC sensitive. In cytarabine-adapted AML cells, increased SAMHD1 and reduced DCK levels contributed to cytarabine and CNDAC resistance., Conclusion: Intrinsic and acquired resistance to CNDAC and related nucleoside analogues are driven by different mechanisms. The lack of cross-resistance between SAMHD1/ DCK substrates and non-substrates provides scope for next-line therapies after treatment failure., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
36. A Potential Role of the CD47/SIRPalpha Axis in COVID-19 Pathogenesis.
- Author
-
McLaughlin KM, Bojkova D, Kandler JD, Bechtel M, Reus P, Le T, Rothweiler F, Wagner JUG, Weigert A, Ciesek S, Wass MN, Michaelis M, and Cinatl J Jr
- Subjects
- Blood Donors, Blotting, Western methods, Bronchi cytology, COVID-19 pathology, COVID-19 virology, Caco-2 Cells, Epithelial Cells metabolism, Epithelial Cells virology, Healthy Volunteers, Humans, Monocytes metabolism, Monocytes virology, Polymerase Chain Reaction methods, RNA, Viral genetics, SARS-CoV-2 genetics, SARS-CoV-2 isolation & purification, Antigens, Differentiation metabolism, CD47 Antigen metabolism, COVID-19 epidemiology, COVID-19 metabolism, Pandemics, Receptors, Immunologic metabolism, SARS-CoV-2 metabolism, Severity of Illness Index, Signal Transduction immunology
- Abstract
The coronavirus SARS-CoV-2 is the cause of the ongoing COVID-19 pandemic. Most SARS-CoV-2 infections are mild or even asymptomatic. However, a small fraction of infected individuals develops severe, life-threatening disease, which is caused by an uncontrolled immune response resulting in hyperinflammation. However, the factors predisposing individuals to severe disease remain poorly understood. Here, we show that levels of CD47, which is known to mediate immune escape in cancer and virus-infected cells, are elevated in SARS-CoV-2-infected Caco-2 cells, Calu-3 cells, and air-liquid interface cultures of primary human bronchial epithelial cells. Moreover, SARS-CoV-2 infection increases SIRPalpha levels, the binding partner of CD47, on primary human monocytes. Systematic literature searches further indicated that known risk factors such as older age and diabetes are associated with increased CD47 levels. High CD47 levels contribute to vascular disease, vasoconstriction, and hypertension, conditions that may predispose SARS-CoV-2-infected individuals to COVID-19-related complications such as pulmonary hypertension, lung fibrosis, myocardial injury, stroke, and acute kidney injury. Hence, age-related and virus-induced CD47 expression is a candidate mechanism potentially contributing to severe COVID-19, as well as a therapeutic target, which may be addressed by antibodies and small molecules. Further research will be needed to investigate the potential involvement of CD47 and SIRPalpha in COVID-19 pathology. Our data should encourage other research groups to consider the potential relevance of the CD47/ SIRPalpha axis in their COVID-19 research.
- Published
- 2021
- Full Text
- View/download PDF
37. Enisamium Inhibits SARS-CoV-2 RNA Synthesis.
- Author
-
Elli S, Bojkova D, Bechtel M, Vial T, Boltz D, Muzzio M, Peng X, Sala F, Cosentino C, Goy A, Guerrini M, Müller L, Cinatl J, Margitich V, and Te Velthuis AJW
- Abstract
Pandemic SARS-CoV-2 causes a mild to severe respiratory disease called coronavirus disease 2019 (COVID-19). While control of the SARS-CoV-2 spread partly depends on vaccine-induced or naturally acquired protective herd immunity, antiviral strategies are still needed to manage COVID-19. Enisamium is an inhibitor of influenza A and B viruses in cell culture and clinically approved in countries of the Commonwealth of Independent States. In vitro, enisamium acts through metabolite VR17-04 and inhibits the activity of the influenza A virus RNA polymerase. Here we show that enisamium can inhibit coronavirus infections in NHBE and Caco-2 cells, and the activity of the SARS-CoV-2 RNA polymerase in vitro. Docking and molecular dynamics simulations provide insight into the mechanism of action and indicate that enisamium metabolite VR17-04 prevents GTP and UTP incorporation. Overall, these results suggest that enisamium is an inhibitor of SARS-CoV-2 RNA synthesis in vitro.
- Published
- 2021
- Full Text
- View/download PDF
38. Differentially conserved amino acid positions may reflect differences in SARS-CoV-2 and SARS-CoV behaviour.
- Author
-
Bojkova D, McGreig JE, McLaughlin KM, Masterson SG, Antczak M, Widera M, Krähling V, Ciesek S, Wass MN, Michaelis M, and Cinatl J
- Abstract
Motivation: SARS-CoV-2 is a novel coronavirus currently causing a pandemic. Here, we performed a combined in-silico and cell culture comparison of SARS-CoV-2 and the closely related SARS-CoV., Results: Many amino acid positions are differentially conserved between SARS-CoV-2 and SARS-CoV, which reflects the discrepancies in virus behaviour, i.e. more effective human-to-human transmission of SARS-CoV-2 and higher mortality associated with SARS-CoV. Variations in the S protein (mediates virus entry) were associated with differences in its interaction with ACE2 (cellular S receptor) and sensitivity to TMPRSS2 (enables virus entry via S cleavage) inhibition. Anti-ACE2 antibodies more strongly inhibited SARS-CoV than SARS-CoV-2 infection, probably due to a stronger SARS-CoV-2 S-ACE2 affinity relative to SARS-CoV S. Moreover, SARS-CoV-2 and SARS-CoV displayed differences in cell tropism. Cellular ACE2 and TMPRSS2 levels did not indicate susceptibility to SARS-CoV-2. In conclusion, we identified genomic variation between SARS-CoV-2 and SARS-CoV that may reflect the differences in their clinical and biological behaviour., Supplementary Information: Supplementary data are available at Bioinformatics online., (© The Author(s) 2021. Published by Oxford University Press.)
- Published
- 2021
- Full Text
- View/download PDF
39. Characterization of ACE Inhibitors and AT 1 R Antagonists with Regard to Their Effect on ACE2 Expression and Infection with SARS-CoV-2 Using a Caco-2 Cell Model.
- Author
-
Reus P, Schneider AK, Ulshöfer T, Henke M, Bojkova D, Cinatl J, Ciesek S, Geisslinger G, Laux V, Grättinger M, Gribbon P, and Schiffmann S
- Abstract
Blood-pressure-lowering drugs are proposed to foster SARS-CoV-2 infection by pharmacological upregulation of angiotensin-converting enzyme 2 (ACE2), the binding partner of the virus spike (S) protein, located on the surface of the host cells. Conversely, it is postulated that angiotensin-renin system antagonists may prevent lung damage caused by SARS-CoV-2 infection, by reducing angiotensin II levels, which can induce permeability of lung endothelial barrier via its interaction with the AT
1 receptor (AT1 R)., Methods: We have investigated the influence of the ACE inhibitors (lisinopril, captopril) and the AT1 antagonists (telmisartan, olmesartan) on the level of ACE2 mRNA and protein expression as well as their influence on the cytopathic effect of SARS-CoV-2 and on the cell barrier integrity in a Caco-2 cell model., Results: The drugs revealed no effect on ACE2 mRNA and protein expression. ACE inhibitors and AT1 R antagonist olmesartan did not influence the infection rate of SARS-CoV-2 and were unable to prevent the SARS-CoV-2-induced cell barrier disturbance. A concentration of 25 µg/mL telmisartan significantly reduced the virus replication rate., Conclusion: ACE inhibitors and AT1 R antagonist showed neither beneficial nor detrimental effects on SARS-CoV-2-infection and cell barrier integrity in vitro at pharmacologically relevant concentrations.- Published
- 2021
- Full Text
- View/download PDF
40. Evaluation of stability and inactivation methods of SARS-CoV-2 in context of laboratory settings.
- Author
-
Widera M, Westhaus S, Rabenau HF, Hoehl S, Bojkova D, Cinatl J Jr, and Ciesek S
- Subjects
- Animals, Caco-2 Cells, Cell Line, Chlorocebus aethiops, Containment of Biohazards, Humans, RNA, Viral, Real-Time Polymerase Chain Reaction, SARS-CoV-2 physiology, Specimen Handling methods, Time Factors, Vero Cells, Anti-Infective Agents pharmacology, COVID-19 prevention & control, COVID-19 virology, Guanidines pharmacology, SARS-CoV-2 drug effects, Thiocyanates pharmacology, Virus Inactivation
- Abstract
The novel coronavirus SARS-CoV-2 is the causative agent of the acute respiratory disease COVID-19, which has become a global concern due to its rapid spread. Laboratory work with SARS-CoV-2 in a laboratory setting was rated to biosafety level 3 (BSL-3) biocontainment level. However, certain research applications in particular in molecular biology require incomplete denaturation of the proteins, which might cause safety issues handling contaminated samples. In this study, we evaluated lysis buffers that are commonly used in molecular biological laboratories for their ability to inactivate SARS-CoV-2. In addition, viral stability in cell culture media at 4 °C and on display glass and plastic surfaces used in laboratory environment was analyzed. Furthermore, we evaluated chemical and non-chemical inactivation methods including heat inactivation, UV-C light, addition of ethanol, acetone-methanol, and PFA, which might be used as a subsequent inactivation step in the case of insufficient inactivation. We infected susceptible Caco-2 and Vero cells with pre-treated SARS-CoV-2 and determined the tissue culture infection dose 50 (TCID
50 ) using crystal violet staining and microscopy. In addition, lysates of infected cells and virus containing supernatant were subjected to RT-qPCR analysis. We have found that guanidine thiocyanate and most of the tested detergent containing lysis buffers were effective in inactivation of SARS-CoV-2, however, the M-PER lysis buffer containing a proprietary detergent failed to inactivate the virus. In conclusion, careful evaluation of the used inactivation methods is required especially for non-denaturing buffers. Additional inactivation steps might be necessary before removal of lysed viral samples from BSL-3., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
41. Famotidine inhibits toll-like receptor 3-mediated inflammatory signaling in SARS-CoV-2 infection.
- Author
-
Mukherjee R, Bhattacharya A, Bojkova D, Mehdipour AR, Shin D, Khan KS, Hei-Yin Cheung H, Wong KB, Ng WL, Cinatl J, Geurink PP, van der Heden van Noort GJ, Rajalingam K, Ciesek S, Hummer G, and Dikic I
- Subjects
- A549 Cells, Binding Sites, Caco-2 Cells, Chemokine CCL2 metabolism, Coronavirus 3C Proteases metabolism, HeLa Cells, Humans, Interferon Regulatory Factor-3 metabolism, Interleukin-6 metabolism, Molecular Docking Simulation, NF-kappa B metabolism, Protein Binding, SARS-CoV-2 physiology, Signal Transduction, Toll-Like Receptor 3 chemistry, Virus Replication, Famotidine pharmacology, Histamine Antagonists pharmacology, SARS-CoV-2 drug effects, Toll-Like Receptor 3 metabolism
- Abstract
Apart from prevention using vaccinations, the management options for COVID-19 remain limited. In retrospective cohort studies, use of famotidine, a specific oral H2 receptor antagonist (antihistamine), has been associated with reduced risk of intubation and death in patients hospitalized with COVID-19. In a case series, nonhospitalized patients with COVID-19 experienced rapid symptom resolution after taking famotidine, but the molecular basis of these observations remains elusive. Here we show using biochemical, cellular, and functional assays that famotidine has no effect on viral replication or viral protease activity. However, famotidine can affect histamine-induced signaling processes in infected Caco2 cells. Specifically, famotidine treatment inhibits histamine-induced expression of Toll-like receptor 3 (TLR3) in SARS-CoV-2 infected cells and can reduce TLR3-dependent signaling processes that culminate in activation of IRF3 and the NF-κB pathway, subsequently controlling antiviral and inflammatory responses. SARS-CoV-2-infected cells treated with famotidine demonstrate reduced expression levels of the inflammatory mediators CCL-2 and IL6, drivers of the cytokine release syndrome that precipitates poor outcome for patients with COVID-19. Given that pharmacokinetic studies indicate that famotidine can reach concentrations in blood that suffice to antagonize histamine H2 receptors expressed in mast cells, neutrophils, and eosinophils, these observations explain how famotidine may contribute to the reduced histamine-induced inflammation and cytokine release, thereby improving the outcome for patients with COVID-19., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
42. Biochemical characterization of protease activity of Nsp3 from SARS-CoV-2 and its inhibition by nanobodies.
- Author
-
Armstrong LA, Lange SM, Dee Cesare V, Matthews SP, Nirujogi RS, Cole I, Hope A, Cunningham F, Toth R, Mukherjee R, Bojkova D, Gruber F, Gray D, Wyatt PG, Cinatl J, Dikic I, Davies P, and Kulathu Y
- Subjects
- A549 Cells, Antigen-Antibody Complex, Humans, Inhibitory Concentration 50, RNA-Dependent RNA Polymerase immunology, RNA-Dependent RNA Polymerase metabolism, Single-Chain Antibodies immunology, Viral Nonstructural Proteins immunology, Viral Nonstructural Proteins metabolism, Antiviral Agents pharmacology, Protease Inhibitors pharmacology, RNA-Dependent RNA Polymerase antagonists & inhibitors, Single-Chain Antibodies pharmacology, Viral Nonstructural Proteins antagonists & inhibitors
- Abstract
Of the 16 non-structural proteins (Nsps) encoded by SARS CoV-2, Nsp3 is the largest and plays important roles in the viral life cycle. Being a large, multidomain, transmembrane protein, Nsp3 has been the most challenging Nsp to characterize. Encoded within Nsp3 is the papain-like protease domain (PLpro) that cleaves not only the viral polypeptide but also K48-linked polyubiquitin and the ubiquitin-like modifier, ISG15, from host cell proteins. We here compare the interactors of PLpro and Nsp3 and find a largely overlapping interactome. Intriguingly, we find that near full length Nsp3 is a more active protease compared to the minimal catalytic domain of PLpro. Using a MALDI-TOF based assay, we screen 1971 approved clinical compounds and identify five compounds that inhibit PLpro with IC50s in the low micromolar range but showed cross reactivity with other human deubiquitinases and had no significant antiviral activity in cellular SARS-CoV-2 infection assays. We therefore looked for alternative methods to block PLpro activity and engineered competitive nanobodies that bind to PLpro at the substrate binding site with nanomolar affinity thus inhibiting the enzyme. Our work highlights the importance of studying Nsp3 and provides tools and valuable insights to investigate Nsp3 biology during the viral infection cycle., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
43. Increased susceptibility of human endothelial cells to infections by SARS-CoV-2 variants.
- Author
-
Wagner JUG, Bojkova D, Shumliakivska M, Luxán G, Nicin L, Aslan GS, Milting H, Kandler JD, Dendorfer A, Heumueller AW, Fleming I, Bibli SI, Jakobi T, Dieterich C, Zeiher AM, Ciesek S, Cinatl J, and Dimmeler S
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, Calnexin metabolism, Cells, Cultured, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum Stress, Endothelial Cells metabolism, Host-Pathogen Interactions, Humans, Membrane Proteins metabolism, Receptors, Virus metabolism, SARS-CoV-2 genetics, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus metabolism, Endoplasmic Reticulum virology, Endothelial Cells virology, SARS-CoV-2 pathogenicity
- Abstract
Coronavirus disease 2019 (COVID-19) spawned a global health crisis in late 2019 and is caused by the novel coronavirus SARS-CoV-2. SARS-CoV-2 infection can lead to elevated markers of endothelial dysfunction associated with higher risk of mortality. It is unclear whether endothelial dysfunction is caused by direct infection of endothelial cells or is mainly secondary to inflammation. Here, we investigate whether different types of endothelial cells are susceptible to SARS-CoV-2. Human endothelial cells from different vascular beds including umbilical vein endothelial cells, coronary artery endothelial cells (HCAEC), cardiac and lung microvascular endothelial cells, or pulmonary arterial cells were inoculated in vitro with SARS-CoV-2. Viral spike protein was only detected in HCAECs after SARS-CoV-2 infection but not in the other endothelial cells tested. Consistently, only HCAEC expressed the SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2), required for virus infection. Infection with the SARS-CoV-2 variants B.1.1.7, B.1.351, and P.2 resulted in significantly higher levels of viral spike protein. Despite this, no intracellular double-stranded viral RNA was detected and the supernatant did not contain infectious virus. Analysis of the cellular distribution of the spike protein revealed that it co-localized with endosomal calnexin. SARS-CoV-2 infection did induce the ER stress gene EDEM1, which is responsible for clearance of misfolded proteins from the ER. Whereas the wild type of SARS-CoV-2 did not induce cytotoxic or pro-inflammatory effects, the variant B.1.1.7 reduced the HCAEC cell number. Of the different tested endothelial cells, HCAECs showed highest viral uptake but did not promote virus replication. Effects on cell number were only observed after infection with the variant B.1.1.7, suggesting that endothelial protection may be particularly important in patients infected with this variant.
- Published
- 2021
- Full Text
- View/download PDF
44. In vitro activity of itraconazole against SARS-CoV-2.
- Author
-
Van Damme E, De Meyer S, Bojkova D, Ciesek S, Cinatl J, De Jonghe S, Jochmans D, Leyssen P, Buyck C, Neyts J, and Van Loock M
- Subjects
- Adenosine analogs & derivatives, Adenosine Monophosphate pharmacology, Alanine pharmacology, Animals, Caco-2 Cells, Cell Line, Tumor, Chlorocebus aethiops, Drug Repositioning, Humans, Vero Cells, Virus Replication drug effects, Adenosine Monophosphate analogs & derivatives, Alanine analogs & derivatives, Antiviral Agents pharmacology, Furans pharmacology, Itraconazole pharmacology, Pyrroles pharmacology, SARS-CoV-2 drug effects, Triazines pharmacology, COVID-19 Drug Treatment
- Abstract
Although vaccination campaigns are currently being rolled out to prevent coronavirus disease (COVID-19), antivirals will remain an important adjunct to vaccination. Antivirals against coronaviruses do not exist, hence global drug repurposing efforts have been carried out to identify agents that may provide clinical benefit to patients with COVID-19. Itraconazole, an antifungal agent, has been reported to have activity against animal coronaviruses. Using cell-based phenotypic assays, the in vitro antiviral activity of itraconazole and 17-OH itraconazole was assessed against clinical isolates from a German and Belgian patient infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Itraconazole demonstrated antiviral activity in human Caco-2 cells (EC
50 = 2.3 µM; 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay). Similarly, its primary metabolite, 17-OH itraconazole, showed inhibition of SARS-CoV-2 activity (EC50 = 3.6 µM). Remdesivir inhibited viral replication with an EC50 = 0.4 µM. Itraconazole and 17-OH itraconazole resulted in a viral yield reduction in vitro of approximately 2-log10 and approximately 1-log10 , as measured in both Caco-2 cells and VeroE6-eGFP cells, respectively. The viral yield reduction brought about by remdesivir or GS-441524 (parent nucleoside of the antiviral prodrug remdesivir; positive control) was more pronounced, with an approximately 3-log10 drop and >4-log10 drop in Caco-2 cells and VeroE6-eGFP cells, respectively. Itraconazole and 17-OH itraconazole exert in vitro low micromolar activity against SARS-CoV-2. Despite the in vitro antiviral activity, itraconazole did not result in a beneficial effect in hospitalized COVID-19 patients in a clinical study (EudraCT Number: 2020-001243-15)., (© 2021 Wiley Periodicals LLC.)- Published
- 2021
- Full Text
- View/download PDF
45. A method for the rational selection of drug repurposing candidates from multimodal knowledge harmonization.
- Author
-
Schultz B, Zaliani A, Ebeling C, Reinshagen J, Bojkova D, Lage-Rupprecht V, Karki R, Lukassen S, Gadiya Y, Ravindra NG, Das S, Baksi S, Domingo-Fernández D, Lentzen M, Strivens M, Raschka T, Cinatl J, DeLong LN, Gribbon P, Geisslinger G, Ciesek S, van Dijk D, Gardner S, Kodamullil AT, Fröhlich H, Peitsch M, Jacobs M, Hoeng J, Eils R, Claussen C, and Hofmann-Apitius M
- Subjects
- Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate therapeutic use, Alanine analogs & derivatives, Alanine therapeutic use, Combined Modality Therapy, Computational Biology, Drug Synergism, Drug Therapy, Combination, GTP Phosphohydrolases therapeutic use, Humans, Knowledge Bases, Nelfinavir therapeutic use, Pandemics, Raloxifene Hydrochloride therapeutic use, Antiviral Agents therapeutic use, Drug Repositioning methods, SARS-CoV-2 physiology, COVID-19 Drug Treatment
- Abstract
The SARS-CoV-2 pandemic has challenged researchers at a global scale. The scientific community's massive response has resulted in a flood of experiments, analyses, hypotheses, and publications, especially in the field of drug repurposing. However, many of the proposed therapeutic compounds obtained from SARS-CoV-2 specific assays are not in agreement and thus demonstrate the need for a singular source of COVID-19 related information from which a rational selection of drug repurposing candidates can be made. In this paper, we present the COVID-19 PHARMACOME, a comprehensive drug-target-mechanism graph generated from a compilation of 10 separate disease maps and sources of experimental data focused on SARS-CoV-2/COVID-19 pathophysiology. By applying our systematic approach, we were able to predict the synergistic effect of specific drug pairs, such as Remdesivir and Thioguanosine or Nelfinavir and Raloxifene, on SARS-CoV-2 infection. Experimental validation of our results demonstrate that our graph can be used to not only explore the involved mechanistic pathways, but also to identify novel combinations of drug repurposing candidates.
- Published
- 2021
- Full Text
- View/download PDF
46. Human Mesenchymal Stromal Cells Are Resistant to SARS-CoV-2 Infection under Steady-State, Inflammatory Conditions and in the Presence of SARS-CoV-2-Infected Cells.
- Author
-
Schäfer R, Spohn G, Bechtel M, Bojkova D, Baer PC, Kuçi S, Seifried E, Ciesek S, and Cinatl J
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, COVID-19 metabolism, Caco-2 Cells, Cell Line, Tumor, Humans, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Inflammation metabolism, Mesenchymal Stem Cells metabolism, Serine Endopeptidases metabolism, COVID-19 Drug Treatment, COVID-19 virology, Inflammation virology, Mesenchymal Stem Cells virology, SARS-CoV-2 pathogenicity
- Abstract
Previous studies reported on the safety and applicability of mesenchymal stem/stromal cells (MSCs) to ameliorate pulmonary inflammation in acute respiratory distress syndrome (ARDS). Thus, multiple clinical trials assessing the potential of MSCs for COVID-19 treatment are underway. Yet, as SARS-inducing coronaviruses infect stem/progenitor cells, it is unclear whether MSCs could be infected by SARS-CoV-2 upon transplantation to COVID-19 patients. We found that MSCs from bone marrow, amniotic fluid, and adipose tissue carry angiotensin-converting enzyme 2 and transmembrane protease serine subtype 2 at low levels on the cell surface under steady-state and inflammatory conditions. We did not observe SARS-CoV-2 infection or replication in MSCs at steady state under inflammatory conditions, or in direct contact with SARS-CoV-2-infected Caco-2 cells. Further, indoleamine 2,3-dioxygenase 1 production in MSCs was not impaired in the presence of SARS-CoV-2. We show that MSCs are resistant to SARS-CoV-2 infection and retain their immunomodulation potential, supporting their potential applicability for COVID-19 treatment., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
47. A SARS-CoV-2 cytopathicity dataset generated by high-content screening of a large drug repurposing collection.
- Author
-
Ellinger B, Bojkova D, Zaliani A, Cinatl J, Claussen C, Westhaus S, Keminer O, Reinshagen J, Kuzikov M, Wolf M, Geisslinger G, Gribbon P, and Ciesek S
- Subjects
- Benzamidines, COVID-19, Caco-2 Cells, Cetylpyridinium, Drug Evaluation, Preclinical, Esters, Guanidines, Humans, Lopinavir, Mefloquine, Papaverine, Antiviral Agents pharmacology, Drug Repositioning, SARS-CoV-2 drug effects
- Abstract
SARS-CoV-2 is a novel coronavirus responsible for the COVID-19 pandemic, in which acute respiratory infections are associated with high socio-economic burden. We applied high-content screening to a well-defined collection of 5632 compounds including 3488 that have undergone previous clinical investigations across 600 indications. The compounds were screened by microscopy for their ability to inhibit SARS-CoV-2 cytopathicity in the human epithelial colorectal adenocarcinoma cell line, Caco-2. The primary screen identified 258 hits that inhibited cytopathicity by more than 75%, most of which were not previously known to be active against SARS-CoV-2 in vitro. These compounds were tested in an eight-point dose response screen using the same image-based cytopathicity readout. For the 67 most active molecules, cytotoxicity data were generated to confirm activity against SARS-CoV-2. We verified the ability of known inhibitors camostat, nafamostat, lopinavir, mefloquine, papaverine and cetylpyridinium to reduce the cytopathic effects of SARS-CoV-2, providing confidence in the validity of the assay. The high-content screening data are suitable for reanalysis across numerous drug classes and indications and may yield additional insights into SARS-CoV-2 mechanisms and potential therapeutic strategies.
- Published
- 2021
- Full Text
- View/download PDF
48. Enisamium is an inhibitor of the SARS-CoV-2 RNA polymerase and shows improvement of recovery in COVID-19 patients in an interim analysis of a clinical trial.
- Author
-
Holubovska O, Bojkova D, Elli S, Bechtel M, Boltz D, Muzzio M, Peng X, Sala F, Cosentino C, Mironenko A, Milde J, Lebed Y, Stammer H, Goy A, Guerrini M, Mueller L, Cinatl J, Margitich V, and Te Velthuis AJW
- Abstract
Pandemic SARS-CoV-2 causes a mild to severe respiratory disease called Coronavirus Disease 2019 (COVID-19). Control of SARS-CoV-2 spread will depend on vaccine-induced or naturally acquired protective herd immunity. Until then, antiviral strategies are needed to manage COVID-19, but approved antiviral treatments, such as remdesivir, can only be delivered intravenously. Enisamium (laboratory code FAV00A, trade name Amizon®) is an orally active inhibitor of influenza A and B viruses in cell culture and clinically approved in countries of the Commonwealth of Independent States. Here we show that enisamium can inhibit SARS-CoV-2 infections in NHBE and Caco-2 cells. In vitro , the previously identified enisamium metabolite VR17-04 directly inhibits the activity of the SARS-CoV-2 RNA polymerase. Docking and molecular dynamics simulations suggest that VR17-04 prevents GTP and UTP incorporation. To confirm enisamium's antiviral properties, we conducted a double-blind, randomized, placebo-controlled trial in adult, hospitalized COVID-19 patients, which needed medical care either with or without supplementary oxygen. Patients received either enisamium (500 mg per dose) or placebo for 7 days. A pre-planned interim analysis showed in the subgroup of patients needing supplementary oxygen (n = 77) in the enisamium group a mean recovery time of 11.1 days, compared to 13.9 days for the placebo group (log-rank test; p=0.0259). No significant difference was found for all patients (n = 373) or those only needing medical care (n = 296). These results thus suggest that enisamium is an inhibitor of SARS-CoV-2 RNA synthesis and that enisamium treatment shortens the time to recovery for COVID-19 patients needing oxygen., Competing Interests: Conflict of interest V.M. and A.G. are employees of Farmak Public Joint Stock Company, Kiev, Ukraine. Part of this research was funded by Farmak Public Joint Stock Company, Kiev, Ukraine.
- Published
- 2021
- Full Text
- View/download PDF
49. SARS-CoV-2 infects and induces cytotoxic effects in human cardiomyocytes.
- Author
-
Bojkova D, Wagner JUG, Shumliakivska M, Aslan GS, Saleem U, Hansen A, Luxán G, Günther S, Pham MD, Krishnan J, Harter PN, Ermel UH, Frangakis AS, Milting H, Zeiher AM, Klingel K, Cinatl J, Dendorfer A, Eschenhagen T, Tschöpe C, Ciesek S, and Dimmeler S
- Subjects
- Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate pharmacology, Alanine analogs & derivatives, Alanine pharmacology, Angiotensin-Converting Enzyme 2 metabolism, Antiviral Agents pharmacology, COVID-19 metabolism, COVID-19 pathology, Caco-2 Cells, Cathepsins metabolism, Heart Diseases drug therapy, Heart Diseases metabolism, Heart Diseases pathology, Host-Pathogen Interactions, Humans, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Reactive Oxygen Species metabolism, SARS-CoV-2 drug effects, Signal Transduction, COVID-19 Drug Treatment, Apoptosis drug effects, COVID-19 virology, Heart Diseases virology, Myocytes, Cardiac virology, SARS-CoV-2 pathogenicity
- Abstract
Aims: Coronavirus disease 2019 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has emerged as a global pandemic. SARS-CoV-2 infection can lead to elevated markers of cardiac injury associated with higher risk of mortality. It is unclear whether cardiac injury is caused by direct infection of cardiomyocytes or is mainly secondary to lung injury and inflammation. Here, we investigate whether cardiomyocytes are permissive for SARS-CoV-2 infection., Methods and Results: Two strains of SARS-CoV-2 infected human induced pluripotent stem cell-derived cardiomyocytes as demonstrated by detection of intracellular double-stranded viral RNA and viral spike glycoprotein expression. Increasing concentrations of viral RNA are detected in supernatants of infected cardiomyocytes, which induced infections in Caco-2 cell lines, documenting productive infections. SARS-CoV-2 infection and induced cytotoxic and proapoptotic effects associated with it abolished cardiomyocyte beating. RNA sequencing confirmed a transcriptional response to viral infection as demonstrated by the up-regulation of genes associated with pathways related to viral response and interferon signalling, apoptosis, and reactive oxygen stress. SARS-CoV-2 infection and cardiotoxicity was confirmed in a 3D cardiosphere tissue model. Importantly, viral spike protein and viral particles were detected in living human heart slices after infection with SARS-CoV-2. Coronavirus particles were further observed in cardiomyocytes of a patient with coronavirus disease 2019. Infection of induced pluripotent stem cell-derived cardiomyocytes was dependent on cathepsins and angiotensin-converting enzyme 2, and was blocked by remdesivir., Conclusion: This study demonstrates that SARS-CoV-2 infects cardiomyocytes in vitro in an angiotensin-converting enzyme 2- and cathepsin-dependent manner. SARS-CoV-2 infection of cardiomyocytes is inhibited by the antiviral drug remdesivir., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2020. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
50. Papain-like protease regulates SARS-CoV-2 viral spread and innate immunity.
- Author
-
Shin D, Mukherjee R, Grewe D, Bojkova D, Baek K, Bhattacharya A, Schulz L, Widera M, Mehdipour AR, Tascher G, Geurink PP, Wilhelm A, van der Heden van Noort GJ, Ovaa H, Müller S, Knobeloch KP, Rajalingam K, Schulman BA, Cinatl J, Hummer G, Ciesek S, and Dikic I
- Subjects
- Animals, Coronavirus Papain-Like Proteases antagonists & inhibitors, Cytokines chemistry, Cytokines metabolism, Deubiquitinating Enzymes antagonists & inhibitors, Deubiquitinating Enzymes chemistry, Deubiquitinating Enzymes metabolism, Humans, Interferon Regulatory Factor-3 metabolism, Interferons immunology, Interferons metabolism, Mice, Models, Molecular, Molecular Dynamics Simulation, NF-kappa B immunology, NF-kappa B metabolism, Protein Binding, SARS-CoV-2 drug effects, SARS-CoV-2 physiology, Ubiquitination, Ubiquitins chemistry, Ubiquitins metabolism, COVID-19 Drug Treatment, COVID-19 immunology, COVID-19 virology, Coronavirus Papain-Like Proteases chemistry, Coronavirus Papain-Like Proteases metabolism, Immunity, Innate, SARS-CoV-2 enzymology, SARS-CoV-2 immunology
- Abstract
The papain-like protease PLpro is an essential coronavirus enzyme that is required for processing viral polyproteins to generate a functional replicase complex and enable viral spread
1,2 . PLpro is also implicated in cleaving proteinaceous post-translational modifications on host proteins as an evasion mechanism against host antiviral immune responses3-5 . Here we perform biochemical, structural and functional characterization of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PLpro (SCoV2-PLpro) and outline differences with SARS-CoV PLpro (SCoV-PLpro) in regulation of host interferon and NF-κB pathways. SCoV2-PLpro and SCoV-PLpro share 83% sequence identity but exhibit different host substrate preferences; SCoV2-PLpro preferentially cleaves the ubiquitin-like interferon-stimulated gene 15 protein (ISG15), whereas SCoV-PLpro predominantly targets ubiquitin chains. The crystal structure of SCoV2-PLpro in complex with ISG15 reveals distinctive interactions with the amino-terminal ubiquitin-like domain of ISG15, highlighting the high affinity and specificity of these interactions. Furthermore, upon infection, SCoV2-PLpro contributes to the cleavage of ISG15 from interferon responsive factor 3 (IRF3) and attenuates type I interferon responses. Notably, inhibition of SCoV2-PLpro with GRL-0617 impairs the virus-induced cytopathogenic effect, maintains the antiviral interferon pathway and reduces viral replication in infected cells. These results highlight a potential dual therapeutic strategy in which targeting of SCoV2-PLpro can suppress SARS-CoV-2 infection and promote antiviral immunity.- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.