Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-21257971

RESUMO

Heterologous COVID-19 vaccination regimens combining vector- and mRNA-based vaccines are already administered, but data on solicited adverse reactions, immunological responses and elicited protection are limited. We aimed to evaluate the reactogenicity, humoral and cellular immune responses towards different SARS-CoV-2 variants after a heterologous ChAdOx1 nCoV-19 BNT162b2 prime-boost vaccination and analyzed a cohort of 26 individuals aged 25-46 (median 30.5) years that received a ChAdOx1 nCoV-19 prime followed by a BNT162b2 boost after an 8- week interval. Self-reported solicited symptoms after ChAdOx1 nCoV-19 prime were in line with previous reports and less severe after the BNT162b2 boost. Antibody titers increased significantly over time resulting in strong neutralization titers two weeks after the BNT162b2 boost. Neutralizing activity against the prevalent strain B.1.1.7 (Alpha) and immune-evading VOC B.1.351 (Beta) was [~]4-fold higher than in individuals receiving homologous BNT162b2 vaccination. No difference was seen in neutralization of VOI B.1.617 (Kappa). In addition, the heterologous vaccination induced CD4+ and CD8+ T cells reactive to SARS-CoV-2 spike peptides of all analyzed variants; Wuhan-Hu-1, B.1.1.7, B.1.351, and P.1 (Gamma). In conclusion, heterologous ChAdOx1 nCoV-19 / BNT162b2 prime-boost vaccination regimen is not associated with serious adverse events and results in a potent humoral immune response and elicits T cell reactivity. Variants B.1.1.7, B.1.351 and B.1.617.1 are potently neutralized by sera of all participants and reactive T cells recognize spike peptides of all tested variants. These results suggest that this heterologous vaccination regimen is at least as immunogenic and protective as homologous vaccinations.

2.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-360545

RESUMO

Many plant juices, extracts and teas have been shown to possess antiviral activity. We here analyzed the virucidal activity of black chokeberry (Aronia melanocarpa), pomegranate (Punica granatum), and elderberry (Sambucus nigra) juice, as well as green tea (Camellia sinensis) against different respiratory viruses. We found that all tested plant derived products effectively inactivated influenza virus, whereas only chokeberry juice diminished SARS-CoV-2 and vaccinia virus infectivity. None of the products inactivated non-enveloped human adenovirus type 5. Thus, black chokeberry juice exerts virucidal activity against different enveloped viral pathogens under in vitro conditions. Whether application of virucidal juices or green tea as oral rinses may lower viral loads in the oral cavity in vivo remains to be evaluated.

3.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-255935

RESUMO

Interferon-induced transmembrane proteins (IFITMs 1, 2 and 3) are thought to restrict numerous viral pathogens including severe acute respiratory syndrome coronaviruses (SARS-CoVs). However, most evidence comes from single-round pseudovirus infection studies of cells that overexpress IFITMs. Here, we verified that artificial overexpression of IFITMs blocks SARS-CoV-2 infection. Strikingly, however, endogenous IFITM expression was essential for efficient infection of genuine SARS-CoV-2 in human lung cells. Our results indicate that the SARS-CoV-2 Spike protein interacts with IFITMs and hijacks them for efficient viral entry. IFITM proteins were expressed and further induced by interferons in human lung, gut, heart and brain cells. Intriguingly, IFITM-derived peptides and targeting antibodies inhibited SARS-CoV-2 entry and replication in human lung cells, cardiomyocytes and gut organoids. Our results show that IFITM proteins are important cofactors for SARS-CoV-2 infection of human cell types representing in vivo targets for viral transmission, dissemination and pathogenesis and suitable targets for therapeutic approaches.

4.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-183764

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). To identify factors of the respiratory tract that suppress SARS-CoV-2, we screened a peptide/protein library derived from bronchoalveolar lavage, and identified 1-antitrypsin (1-AT) as specific inhibitor of SARS-CoV-2. 1-AT targets the viral spike protein and blocks SARS-CoV-2 infection of human airway epithelium at physiological concentrations. Our findings show that endogenous 1-AT restricts SARS-CoV-2 and repurposes 1-AT-based drugs for COVID-19 therapy.

5.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-155689

RESUMO

SARS-CoV-2 RNA has been detected in the human breast milk of infected mothers, raising concerns regarding the safety of breastfeeding upon infection. We here show that holder pasteurization inactivates SARS-CoV-2 and provides an alternative and safe option for infected mothers to continue feeding breast milk to their infants.

6.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-150862

RESUMO

SARS-CoV-2 is a novel pandemic coronavirus that caused a global health and economic crisis. The development of efficient drugs and vaccines against COVID-19 requires detailed knowledge about SARS-CoV-2 biology. Several techniques to detect SARS-CoV-2 infection have been established, mainly based on counting infected cells by staining plaques or foci, or by quantifying the viral genome by PCR. These methods are laborious, time-consuming and expensive and therefore not suitable for a high sample throughput or rapid diagnostics. We here report a novel enzyme-based immunodetection assay that directly quantifies the amount of de novo synthesized viral spike protein within fixed and permeabilized cells. This in-cell ELISA enables a rapid and quantitative detection of SARS-CoV-2 infection in microtiter format, regardless of the virus isolate or target cell culture. It follows the established method of performing ELISA assays and does not require expensive instrumentation. Utilization of the in-cell ELISA allows to e.g. determine TCID50 of virus stocks, antiviral efficiencies (IC50 values) of drugs or neutralizing activity of sera. Thus, the in-cell spike ELISA represents a promising alternative to study SARS-CoV-2 infection and inhibition and may facilitate future research. HighlightsO_LIDetermination of SARS-CoV-2 infection by enzymatically quantifying the expression of viral spike protein in bulk cell cultures C_LIO_LITargeting a highly conserved region in the S2 subunit of the S protein allows broad detection of several SARS-CoV-2 isolates in different cell lines C_LIO_LIScreening of antivirals in microtiter format and determining the antiviral activity as inhibitory concentrations 50 (IC50) C_LI

7.
Preprint em Inglês | bioRxiv | ID: ppbiorxiv-144816

RESUMO

Gastrointestinal symptoms in COVID-19 are associated with prolonged symptoms and increased severity. We employed human intestinal organoids derived from pluripotent stem cells (PSC-HIOs) to analyze SARS-CoV-2 pathogenesis and to validate efficacy of specific drugs in the gut. Certain, but not all cell types in PSC-HIOs express SARS-CoV-2 entry factors ACE2 and TMPRSS2, rendering them susceptible to SARS-CoV-2 infection. Remdesivir, a promising drug to treat COVID-19, effectively suppressed SARS-CoV-2 infection of PSC-HIOs. In contrast, the histamine-2-blocker famotidine showed no effect. Thus, PSC-HIOs provide an interesting platform to study SARS-CoV-2 infection and to identify or validate drugs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...