13 results on '"Trimpert, J"'
Search Results
2. Key benefits of dexamethasone and antibody treatment in COVID-19 hamster models revealed by single cell transcriptomics
- Author
-
Wyler, E., Adler, J.M., Eschke, K., Teixeira Alves, G., Peidli, S., Pott, F., Kazmierski, J., Michalick, L., Kershaw, O., Bushe, J., Andreotti, S., Pennitz, P., Abdelgawad, A., Postmus, D., Goffinet, C., Kreye, J., Reincke, S.M., Prüss, H., Blüthgen, N., Gruber, A.D., Kuebler, W.M., Witzenrath, M., Landthaler, M., Nouailles, G., and Trimpert, J.
- Subjects
Cancer Research - Abstract
For COVID-19, effective and well-understood treatment options are still scarce. Since vaccine efficacy is challenged by novel variants, short-lasting immunity and vaccine hesitancy, understanding and optimizing therapeutic options remains essential. We aimed at better understanding the effects of two standard-of-care drugs, dexamethasone and anti-SARS-CoV-2 antibodies, on infection and host responses. By using two COVID-19 hamster models, pulmonary immune responses were analyzed to characterize effects of single or combinatorial treatments. Pulmonary viral burden was reduced by anti-SARS-CoV-2 antibody treatment, and similar or increased by dexamethasone alone. Dexamethasone exhibited strong anti-inflammatory effects and prevented fulminant disease in a severe disease model. Combination therapy showed additive benefits with both anti-viral and anti-inflammatory potency. Bulk and single-cell transcriptomic analyses confirmed dampened inflammatory cell recruitment into lungs upon dexamethasone treatment, and identified a specifically responsive subpopulation of neutrophils, thereby indicating a potential mechanism of action. Our analyses confirm the anti-inflammatory properties of dexamethasone and suggest possible mechanisms, validate anti-viral effects of anti-SARS-CoV-2 antibody treatment, and reveal synergistic effects of a combination therapy, thus informing more effective COVID-19 therapies.
- Published
- 2022
3. A pulmonologist's guide to perform and analyse cross-species single lung cell transcriptomics
- Author
-
Pennitz, P., Kirsten, H., Friedrich, V.D., Wyler, E., Goekeri, C., Obermayer, B., Heinz, G.A., Mashreghi, M.F., Büttner, M., Trimpert, J., Landthaler, M., Suttorp, N., Hocke, A.C., Hippenstiel, S., Tönnies, M., Scholz, M., Kuebler, W.M., Witzenrath, M., Hoenzke, K., and Nouailles, G.
- Subjects
Single lung cell transcriptomics ,Cancer Research ,Single-cell RNA sequencing (scRNA-seq) ,Base Sequence ,Swine ,Rats ,Mice ,Pulmonologists ,Species Specificity ,Cricetinae ,Chlorocebus aethiops ,Pulmonologist’s guide ,Animals ,Humans ,ddc:610 ,Technology Platforms ,Transcriptome ,Lung ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit::610 Medizin und Gesundheit - Abstract
Single-cell ribonucleic acid sequencing is becoming widely employed to study biological processes at a novel resolution depth. The ability to analyse transcriptomes of multiple heterogeneous cell types in parallel is especially valuable for cell-focused lung research where a variety of resident and recruited cells are essential for maintaining organ functionality. We compared the single-cell transcriptomes from publicly available and unpublished datasets of the lungs in six different species: human (Homo sapiens), African green monkey (Chlorocebus sabaeus), pig (Sus domesticus), hamster (Mesocricetus auratus), rat (Rattus norvegicus) and mouse (Mus musculus) by employing RNA velocity and intercellular communication based on ligand-receptor co-expression, among other techniques. Specifically, we demonstrated a workflow for interspecies data integration, applied a single unified gene nomenclature, performed cell-specific clustering and identified marker genes for each species. Overall, integrative approaches combining newly sequenced as well as publicly available datasets could help identify species-specific transcriptomic signatures in both healthy and diseased lung tissue and select appropriate models for future respiratory research.
- Published
- 2022
- Full Text
- View/download PDF
4. Virus-induced senescence is driver and therapeutic target in COVID-19
- Author
-
Lee, S., Yu, Y., Trimpert, J., Benthani, F., Mairhofer, M., Richter-Pechanska, P., Wyler, E., Belenki, D., Kaltenbrunner, S., Pammer, M., Kausche, L., Firsching, T.C., Dietert, K., Schotsaert, M., Martínez-Romero, C., Singh, G., Kunz, S., Niemeyer, D., Ghanem, R., Salzer, H.J.F., Paar, C., Mülleder, M., Uccellini, M., Michaelis, E.G., Khan, A., Lau, A., Schönlein, M., Habringer, A., Tomasits, J., Adler, J.M., Kimeswenger, S., Gruber, A.D., Hoetzenecker, W., Steinkellner, H., Purfuerst, B., Motz, R., Di Pierro, F., Lamprecht, B., Osterrieder, N., Landthaler, M., Drosten, C., García-Sastre, A., Langer, R., Ralser, M., Eils, R., Reimann, M., Fan, D.N.Y., and Schmitt, C.A.
- Subjects
Cancer Research ,Technology Platforms - Abstract
Derailed cytokine and immune cell networks account for organ damage and clinical severity of COVID-19. Here we show that SARS-CoV-2, like other viruses, evokes cellular senescence as a primary stress response in infected cells. Virus-induced senescence (VIS) is indistinguishable from other forms of cellular senescence and accompanied by a senescence-associated secretory phenotype (SASP), composed of pro-inflammatory cytokines, extracellular matrix-active factors and pro-coagulatory mediators. COVID-19 patients displayed markers of senescence in their airway mucosa in situ and elevated serum levels of SASP factors. Mirroring COVID-19 hallmark features such as macrophage and neutrophil infiltration, endothelial damage and widespread thrombosis in affected lung tissue in vitro assays demonstrated macrophage activation with SASP-reminiscent secretion, complement lysis and SASP-amplifying secondary senescence of endothelial cells, neutrophil extracellular trap (NET) formation as well as activation of platelets and the clotting cascade in response to supernatant of VIS cells, including SARS-CoV-2-induced senescence. Senolytics such as Navitoclax and Dasatinib/Quercetin selectively eliminated VIS cells, mitigated COVID-19-reminiscent lung disease and reduced inflammation in SARS-CoV-2-driven hamster and mouse models. Our findings mark VIS as pathogenic trigger of COVID-19-related cytokine escalation and organ damage, and suggest senolytic targeting of virus-infected cells as a novel treatment option against SARS-CoV-2 and perhaps other viral infections.
- Published
- 2021
5. Epithelial response to IFN-γ promotes SARS-CoV-2 infection
- Author
-
Heuberger, J., Trimpert, J., Vladimirova, D., Goosmann, C., Lin, M., Schmuck, R., Mollenkopf, H.J., Brinkmann, V., Tacke, F., Osterrieder, N., and Sigal, M.
- Subjects
Cancer Research ,hormones, hormone substitutes, and hormone antagonists - Abstract
SARS-CoV-2, the agent that causes COVID-19, invades epithelial cells, including those of the respiratory and gastrointestinal mucosa, using angiotensin-converting enzyme-2 (ACE2) as a receptor. Subsequent inflammation can promote rapid virus clearance, but severe cases of COVID-19 are characterized by an inefficient immune response that fails to clear the infection. Using primary epithelial organoids from human colon, we explored how the central antiviral mediator IFN-γ, which is elevated in COVID-19, affects epithelial cell differentiation, ACE2 expression, and susceptibility to infection with SARS-CoV-2. In mouse and human colon, ACE2 is mainly expressed by surface enterocytes. Inducing enterocyte differentiation in organoid culture resulted in increased ACE2 production. IFN-γ treatment promoted differentiation into mature KRT20+ enterocytes expressing high levels of ACE2, increased susceptibility to SARS-CoV-2 infection and resulted in enhanced virus production in infected cells. Similarly, infection-induced epithelial interferon signaling promoted enterocyte maturation and enhanced ACE2 expression. We here reveal a mechanism by which IFN-γ-driven inflammatory responses induce a vulnerable epithelial state with robust replication of SARS-CoV-2, which may have an impact on disease outcome and virus transmission.
- Published
- 2021
6. Epithelial response to IFN-c promotes SARS-CoV-2 infection
- Author
-
Heuberger, J., Trimpert, J., Vladimirova, D., Goosmann, C., Lin, M., Schmuck, R., Mollenkopf, H., Brinkmann, V., Tacke, F., Osterrieder, N., and Sigal, M.
- Published
- 2021
7. ICTV Virus Taxonomy Profile: Herpesviridae 2021
- Author
-
Gatherer, D, Depledge, DP, Hartley, CA, Szpara, ML, Vaz, PK, Benko, M, Brandt, CR, Bryant, NA, Dastjerdi, A, Doszpoly, A, Gompels, UA, Inoue, N, Jarosinski, KW, Kaul, R, Lacoste, V, Norberg, P, Origgi, FC, Orton, RJ, Pellett, PE, Schmid, DS, Spatz, SJ, Stewart, JP, Trimpert, J, Waltzek, TB, Davison, AJ, Gatherer, D, Depledge, DP, Hartley, CA, Szpara, ML, Vaz, PK, Benko, M, Brandt, CR, Bryant, NA, Dastjerdi, A, Doszpoly, A, Gompels, UA, Inoue, N, Jarosinski, KW, Kaul, R, Lacoste, V, Norberg, P, Origgi, FC, Orton, RJ, Pellett, PE, Schmid, DS, Spatz, SJ, Stewart, JP, Trimpert, J, Waltzek, TB, and Davison, AJ
- Abstract
Members of the family Herpesviridae have enveloped, spherical virions with characteristic complex structures consisting of symmetrical and non-symmetrical components. The linear, double-stranded DNA genomes of 125-241 kbp contain 70-170 genes, of which 43 have been inherited from an ancestral herpesvirus. In general, herpesviruses have coevolved with and are highly adapted to their hosts, which comprise many mammalian, avian and reptilian species. Following primary infection, they are able to establish lifelong latent infection, during which there is limited viral gene expression. Severe disease is usually observed only in the foetus, the very young, the immunocompromised or following infection of an alternative host. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Herpesviridae, which is available at ictv.global/report/herpesviridae.
- Published
- 2021
8. Fatal Elephant Endotheliotropic Herpesvirus Infection of Two Young Asian Elephants
- Author
-
Pavulraj, S, Eschke, K, Prahl, A, Flugger, M, Trimpert, J, van den Doel, Petra, Andreotti, S, Kaessmeyer, S, Osterrieder, N, Azab, W, Pavulraj, S, Eschke, K, Prahl, A, Flugger, M, Trimpert, J, van den Doel, Petra, Andreotti, S, Kaessmeyer, S, Osterrieder, N, and Azab, W
- Published
- 2019
9. The FDA-Approved Drug Cobicistat Synergizes with Remdesivir to Inhibit SARS-CoV-2 Replication in Vitro and Decreases Viral Titers and Disease Progression in Syrian Hamsters
- Author
-
Iart Luca Shytaj, Mohamed Fares, Lara Gallucci, Bojana Lucic, Mahmoud M. Tolba, Liv Zimmermann, Julia M. Adler, Na Xing, Judith Bushe, Achim D. Gruber, Ina Ambiel, Ahmed Taha Ayoub, Mirko Cortese, Christopher J. Neufeldt, Bettina Stolp, Mohamed Hossam Sobhy, Moustafa Fathy, Min Zhao, Vibor Laketa, Ricardo Sobhie Diaz, Richard E. Sutton, Petr Chlanda, Steeve Boulant, Ralf Bartenschlager, Megan L. Stanifer, Oliver T. Fackler, Jakob Trimpert, Andrea Savarino, Marina Lusic, Shytaj, I. L., Fares, M., Gallucci, L., Lucic, B., Tolba, M. M., Zimmermann, L., Adler, J. M., Xing, N., Bushe, J., Gruber, A. D., Ambiel, I., Ayoub, A. T., Cortese, M., Neufeldt, C. J., Stolp, B., Sobhy, M. H., Fathy, M., Zhao, M., Laketa, V., Diaz, R. S., Sutton, R. E., Chlanda, P., Boulant, S., Bartenschlager, R., Stanifer, M. L., Fackler, O. T., Trimpert, J., Savarino, A., and Lusic, M.
- Subjects
direct-acting antiviral ,Mesocricetus ,drug repurposing ,SARS-CoV-2 ,COVID-19 ,remdesivir ,Alanine/analogs & derivatives ,Antiviral Agents/pharmacology ,Hepatitis C, Chronic ,Viral Load ,cobicistat ,spike protein ,COVID-19/drug therapy ,Microbiology ,Adenosine Monophosphate/analogs & derivatives ,Cricetinae ,Virology ,Disease Progression ,Animals ,Humans ,Cobicistat ,Pandemics - Abstract
The lack of effective antiviral treatments against SARS-CoV-2 is a significant limitation in the fight against the COVID-19 pandemic. Single-drug regimens have so far yielded limited results, indicating that combinations of antivirals might be required, as previously seen for other RNA viruses.Combinations of direct-acting antivirals are needed to minimize drug resistance mutations and stably suppress replication of RNA viruses. Currently, there are limited therapeutic options against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and testing of a number of drug regimens has led to conflicting results. Here, we show that cobicistat, which is an FDA-approved drug booster that blocks the activity of the drug-metabolizing proteins cytochrome P450-3As (CYP3As) and P-glycoprotein (P-gp), inhibits SARS-CoV-2 replication. Two independent cell-to-cell membrane fusion assays showed that the antiviral effect of cobicistat is exerted through inhibition of spike protein-mediated membrane fusion. In line with this, incubation with low-micromolar concentrations of cobicistat decreased viral replication in three different cell lines including cells of lung and gut origin. When cobicistat was used in combination with remdesivir, a synergistic effect on the inhibition of viral replication was observed in cell lines and in a primary human colon organoid. This was consistent with the effects of cobicistat on two of its known targets, CYP3A4 and P-gp, the silencing of which boosted the in vitro antiviral activity of remdesivir in a cobicistat-like manner. When administered in vivo to Syrian hamsters at a high dose, cobicistat decreased viral load and mitigated clinical progression. These data highlight cobicistat as a therapeutic candidate for treating SARS-CoV-2 infection and as a potential building block of combination therapies for COVID-19. IMPORTANCE The lack of effective antiviral treatments against SARS-CoV-2 is a significant limitation in the fight against the COVID-19 pandemic. Single-drug regimens have so far yielded limited results, indicating that combinations of antivirals might be required, as previously seen for other RNA viruses. Our work introduces the drug booster cobicistat, which is approved by the FDA and typically used to potentiate the effect of anti-HIV protease inhibitors, as a candidate inhibitor of SARS-CoV-2 replication. Beyond its direct activity as an antiviral, we show that cobicistat can enhance the effect of remdesivir, which was one of the first drugs proposed for treatment of SARS-CoV-2. Overall, the dual action of cobicistat as a direct antiviral and a drug booster can provide a new approach to design combination therapies and rescue the activity of compounds that are only partially effective in monotherapy.
- Published
- 2022
- Full Text
- View/download PDF
10. Author Correction: Virus-induced senescence is a driver and therapeutic target in COVID-19.
- Author
-
Lee S, Yu Y, Trimpert J, Benthani F, Mairhofer M, Richter-Pechanska P, Wyler E, Belenki D, Kaltenbrunner S, Pammer M, Kausche L, Firsching TC, Dietert K, Schotsaert M, Martínez-Romero C, Singh G, Kunz S, Niemeyer D, Ghanem R, Salzer HJF, Paar C, Mülleder M, Uccellini M, Michaelis EG, Khan A, Lau A, Schönlein M, Habringer A, Tomasits J, Adler JM, Kimeswenger S, Gruber AD, Hoetzenecker W, Steinkellner H, Purfürst B, Motz R, Di Pierro F, Lamprecht B, Osterrieder N, Landthaler M, Drosten C, García-Sastre A, Langer R, Ralser M, Eils R, Reimann M, Fan DNY, and Schmitt CA
- Published
- 2025
- Full Text
- View/download PDF
11. A systematic analysis of anthocyanins inhibiting human, murine, and equine herpesviruses.
- Author
-
Roll V, Diesendorf V, Roewer N, Abdelgawad A, Roewer J, Trimpert J, and Bodem J
- Subjects
- Child, Humans, Animals, Horses, Mice, Anthocyanins pharmacology, Anthocyanins analysis, Antiviral Agents pharmacology, Plant Extracts pharmacology, Mammals, Hepatitis C, Chronic, Herpesvirus 1, Human, Elaeocarpaceae, Cytomegalovirus Infections
- Abstract
Background: Herpesviruses are common animal and human pathogens that cause severe health problems in children, immunocompromised patients, and infected animals with a host range from fish to mammals. Anthocyanin-containing plant extracts have been described as potent antivirals, which might cause fewer harmful side effects than direct-acting antivirals. Here, we report that an extract of Aristotelia chilensis (Molina) Stuntz (Elaeocarpaceae) (MBE) with a high content of the anthocyanin delphinidin suppresses lytic replication of equine, murine and human herpesviruses of replication in vitro., Methods: We treated cultured cells with MBE and purified individual anthocyanins present in the extract to determine the most active compound at different concentrations. We subsequently infected the cultures with human herpesviruses 1 (HSV-1) or 8 (HHV-8), murine cytomegalovirus (CMV), or equine herpesviruses 1 (EHV-1) and determined the number of infected cells and viral infectivity., Results: MBE inhibited the HSV-1, murine CMV, and EHV-1 by up to 2 orders of magnitude. In the presence of the stabilizing randomly methylated-beta-cyclodextrin, the inhibitory concentration could be lowered significantly. We identified delphinidin as an active antiviral compound and showed that the non-glycosylated delphinidin solved and stabilized with sulfobutylether-beta-cyclodextrin allowed usage of approximately 50 times lower concentrations., Conclusion: Glycosylated delphinidin derivatives were identified as active antiviral compounds of MBE. This suggests that plant extracts rich in delphinidin-anthocyanins have potent antiviral properties that could be used in treatment and prevention., 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 Elsevier GmbH. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. The trispecific DARPin ensovibep inhibits diverse SARS-CoV-2 variants.
- Author
-
Rothenberger S, Hurdiss DL, Walser M, Malvezzi F, Mayor J, Ryter S, Moreno H, Liechti N, Bosshart A, Iss C, Calabro V, Cornelius A, Hospodarsch T, Neculcea A, Looser T, Schlegel A, Fontaine S, Villemagne D, Paladino M, Schiegg D, Mangold S, Reichen C, Radom F, Kaufmann Y, Schaible D, Schlegel I, Zitt C, Sigrist G, Straumann M, Wolter J, Comby M, Sacarcelik F, Drulyte I, Lyoo H, Wang C, Li W, Du W, Binz HK, Herrup R, Lusvarghi S, Neerukonda SN, Vassell R, Wang W, Adler JM, Eschke K, Nascimento M, Abdelgawad A, Gruber AD, Bushe J, Kershaw O, Knutson CG, Balavenkatraman KK, Ramanathan K, Wyler E, Teixeira Alves LG, Lewis S, Watson R, Haeuptle MA, Zürcher A, Dawson KM, Steiner D, Weiss CD, Amstutz P, van Kuppeveld FJM, Stumpp MT, Bosch BJ, Engler O, and Trimpert J
- Subjects
- Animals, Cricetinae, Humans, Designed Ankyrin Repeat Proteins, Cryoelectron Microscopy, Antibodies, Monoclonal therapeutic use, Combined Antibody Therapeutics, Antibodies, Neutralizing, SARS-CoV-2 genetics, COVID-19
- Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with potential resistance to existing drugs emphasizes the need for new therapeutic modalities with broad variant activity. Here we show that ensovibep, a trispecific DARPin (designed ankyrin repeat protein) clinical candidate, can engage the three units of the spike protein trimer of SARS-CoV-2 and inhibit ACE2 binding with high potency, as revealed by cryo-electron microscopy analysis. The cooperative binding together with the complementarity of the three DARPin modules enable ensovibep to inhibit frequent SARS-CoV-2 variants, including Omicron sublineages BA.1 and BA.2. In Roborovski dwarf hamsters infected with SARS-CoV-2, ensovibep reduced fatality similarly to a standard-of-care monoclonal antibody (mAb) cocktail. When used as a single agent in viral passaging experiments in vitro, ensovibep reduced the emergence of escape mutations in a similar fashion to the same mAb cocktail. These results support further clinical evaluation of ensovibep as a broad variant alternative to existing targeted therapies for Coronavirus Disease 2019 (COVID-19)., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
13. Virus-induced senescence is a driver and therapeutic target in COVID-19.
- Author
-
Lee S, Yu Y, Trimpert J, Benthani F, Mairhofer M, Richter-Pechanska P, Wyler E, Belenki D, Kaltenbrunner S, Pammer M, Kausche L, Firsching TC, Dietert K, Schotsaert M, Martínez-Romero C, Singh G, Kunz S, Niemeyer D, Ghanem R, Salzer HJF, Paar C, Mülleder M, Uccellini M, Michaelis EG, Khan A, Lau A, Schönlein M, Habringer A, Tomasits J, Adler JM, Kimeswenger S, Gruber AD, Hoetzenecker W, Steinkellner H, Purfürst B, Motz R, Di Pierro F, Lamprecht B, Osterrieder N, Landthaler M, Drosten C, García-Sastre A, Langer R, Ralser M, Eils R, Reimann M, Fan DNY, and Schmitt CA
- Subjects
- Aniline Compounds pharmacology, Aniline Compounds therapeutic use, Animals, COVID-19 complications, Cell Line, Cricetinae, Dasatinib pharmacology, Dasatinib therapeutic use, Disease Models, Animal, Female, Humans, Male, Mice, Quercetin pharmacology, Quercetin therapeutic use, SARS-CoV-2 drug effects, Sulfonamides pharmacology, Sulfonamides therapeutic use, Thrombosis complications, Thrombosis immunology, Thrombosis metabolism, COVID-19 pathology, COVID-19 virology, Cellular Senescence drug effects, Molecular Targeted Therapy, SARS-CoV-2 pathogenicity, COVID-19 Drug Treatment
- Abstract
Derailed cytokine and immune cell networks account for the organ damage and the clinical severity of COVID-19 (refs.
1-4 ). Here we show that SARS-CoV-2, like other viruses, evokes cellular senescence as a primary stress response in infected cells. Virus-induced senescence (VIS) is indistinguishable from other forms of cellular senescence and is accompanied by a senescence-associated secretory phenotype (SASP), which comprises pro-inflammatory cytokines, extracellular-matrix-active factors and pro-coagulatory mediators5-7 . Patients with COVID-19 displayed markers of senescence in their airway mucosa in situ and increased serum levels of SASP factors. In vitro assays demonstrated macrophage activation with SASP-reminiscent secretion, complement lysis and SASP-amplifying secondary senescence of endothelial cells, which mirrored hallmark features of COVID-19 such as macrophage and neutrophil infiltration, endothelial damage and widespread thrombosis in affected lung tissue1,8,9 . Moreover, supernatant from VIS cells, including SARS-CoV-2-induced senescence, induced neutrophil extracellular trap formation and activation of platelets and the clotting cascade. Senolytics such as navitoclax and a combination of dasatinib plus quercetin selectively eliminated VIS cells, mitigated COVID-19-reminiscent lung disease and reduced inflammation in SARS-CoV-2-infected hamsters and mice. Our findings mark VIS as a pathogenic trigger of COVID-19-related cytokine escalation and organ damage, and suggest that senolytic targeting of virus-infected cells is a treatment option against SARS-CoV-2 and perhaps other viral infections., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.