Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Wibke Bayer is active.

Publication


Featured researches published by Wibke Bayer.


Journal of Virology | 2010

Vaccination with an Adenoviral Vector That Encodes and Displays a Retroviral Antigen Induces Improved Neutralizing Antibody and CD4+ T-Cell Responses and Confers Enhanced Protection

Wibke Bayer; Matthias Tenbusch; Ruth Lietz; Lena Johrden; Simone Schimmer; Klaus Überla; Ulf Dittmer; Oliver Wildner

ABSTRACT We present a new type of adenoviral vector that both encodes and displays a vaccine antigen on the capsid, thus combining in itself gene-based and protein vaccination; this vector resulted in an improved vaccination outcome in the Friend virus (FV) model. For presentation of the envelope protein gp70 of Friend murine leukemia virus on the adenoviral capsid, gp70 was fused to the adenovirus capsid protein IX. When compared to vaccination with conventional FV Env- and Gag-encoding adenoviral vectors, vaccination with the adenoviral vector that encodes and displays pIX-gp70 combined with an FV Gag-encoding vector resulted in significantly improved protection against systemic FV challenge infection, with highly controlled viral loads in plasma and spleen. This improved protection correlated with improved neutralizing antibody titers and stronger CD4+ T-cell responses. Using a vector that displays gp70 without encoding it, we found that while the antigen display on the capsid alone was sufficient to induce high levels of binding antibodies, in vivo expression was necessary for the induction of neutralizing antibodies. This new type of adenovirus-based vaccine could be a valuable tool for vaccination.


Nanomedicine: Nanotechnology, Biology and Medicine | 2014

Prophylactic and therapeutic vaccination with a nanoparticle-based peptide vaccine induces efficient protective immunity during acute and chronic retroviral infection.

Torben Knuschke; Wibke Bayer; Olga Rotan; Viktoriya Sokolova; Munisch Wadwa; Carsten J. Kirschning; Wiebke Hansen; Ulf Dittmer; Matthias Epple; Jan Buer; Astrid M. Westendorf

UNLABELLED Retroviral infections e.g. HIV still represent a unique burden in the field of vaccine research. A common challenge in vaccine design is to find formulations that create appropriate immune responses to protect against and/or control the given pathogen. Nanoparticles have been considered to be ideal vaccination vehicles that mimic invading pathogens. In this study, we present biodegradable calcium phosphate (CaP) nanoparticles, functionalized with CpG and retroviral T cell epitopes of Friend virus (FV) as excellent vaccine delivery system. CaP nanoparticles strongly increased antigen delivery to antigen-presenting cells to elicit a highly efficient T cell-mediated immune response against retroviral FV infection. Moreover, single-shot immunization of chronically FV-infected mice with functionalized CaP nanoparticles efficiently reactivated effector T cells which led to a significant decrease in viral loads. Thus, our findings clearly indicate that a nanoparticle-based peptide immunization is a promising approach to improve antiretroviral vaccination. FROM THE CLINICAL EDITOR In this study, biodegradable calcium phosphate nanoparticles were used as a vaccine delivery system after functionalization with CpG and Friend virus-derived T-cell epitopes. This vaccination strategy resulted in increased T-cell mediated immune response even in chronically infected mice, providing a promising approach to the development of clinically useful antiretroviral vaccination strategies.


Retrovirology | 2011

Improved vaccine protection against retrovirus infection after co-administration of adenoviral vectors encoding viral antigens and type I interferon subtypes

Wibke Bayer; Ruth Lietz; Teona Ontikatze; Lena Johrden; Matthias Tenbusch; Ghulam Nabi; Simone Schimmer; Peter Groitl; Hans Wolf; Cassandra M. Berry; Klaus Überla; Ulf Dittmer; Oliver Wildner

BackgroundType I interferons (IFNs) exhibit direct antiviral effects, but also distinct immunomodulatory properties. In this study, we analyzed type I IFN subtypes for their effect on prophylactic adenovirus-based anti-retroviral vaccination of mice against Friend retrovirus (FV) or HIV.ResultsMice were vaccinated with adenoviral vectors encoding FV Env and Gag proteins alone or in combination with vectors encoding IFNα1, IFNα2, IFNα4, IFNα5, IFNα6, IFNα9 or IFNβ. Only the co-administration of adenoviral vectors encoding IFNα2, IFNα4, IFNα6 and IFNα9 resulted in strongly improved immune protection of vaccinated mice from subsequent FV challenge infection with high control over FV-induced splenomegaly and reduced viral loads. The level of protection correlated with augmented virus-specific CD4+ T cell responses and enhanced antibody titers. Similar results were obtained when mice were vaccinated against HIV with adenoviral vectors encoding HIV Env and Gag-Pol in combination with various type I IFN encoding vectors. Here mainly CD4+ T cell responses were enhanced by IFNα subtypes.ConclusionsOur results indicate that certain IFNα subtypes have the potential to improve the protective effect of adenovirus-based vaccines against retroviruses. This correlated with augmented virus-specific CD4+ T cell and antibody responses. Thus, co-expression of select type I IFNs may be a valuable tool for the development of anti-retroviral vaccines.


Molecular Cancer Therapeutics | 2007

Intratumoral expression of respiratory syncytial virus fusion protein in combination with cytokines encoded by adenoviral vectors as in situ tumor vaccine for colorectal cancer

Dennis Hoffmann; Wibke Bayer; Thomas Grunwald; Oliver Wildner

Although cancers can naturally elicit immune responses, immune ignorance is a common observation preventing immune-mediated elimination of tumor cells. We assessed whether intratumoral expression of respiratory syncytial virus fusion (RSV-F) protein, encoded by a replication-defective adenovirus vector (Ad.RSV-F), alone or in combination with local coexpression of cytokines can induce tumor-specific immune responses in a syngeneic murine colon cancer model. We confirmed in vitro by dye colocalization that transduction of murine cells with Ad.RSV-F induces cell-cell fusion. In vivo, we showed in a bilateral syngeneic s.c. colon cancer model in C57BL/6 and BALB/c mice that intratumoral injection of Ad.RSV-F leads to a significant volume reduction not only of the directly vector-treated tumor but also of the contralateral not directly vector-treated tumor. The intratumoral administration of Ad.RSV-F in combination with adenovirus vectors encoding interleukin (IL)-2, IL-12, IL-18, IL-21, or granulocyte macrophage colony-stimulating factor significantly enhanced the antitumor effect on the directly vector-treated tumor and also on the contralateral tumor. The antineoplastic efficacy of this combined treatment was significantly higher than that of the individual treatment components and was associated with the induction of a tumor-specific CTL response and increased infiltration of the tumors by natural killer cells and macrophages. Intratumoral coexpression of RSV-F and IL-21 resulted in the highest tumor growth inhibition and improved survival. Our experimental data indicate that intratumoral expression of RSV-F in combination with cytokines is a promising novel tool for the development of in situ tumor vaccination approaches. [Mol Cancer Ther 2007;6(7):1942–50]


Retrovirology | 2011

Distinct roles of CD4+ T cell subpopulations in retroviral immunity: lessons from the Friend virus mouse model.

Savita Nair; Wibke Bayer; Mickaël J.-Y. Ploquin; George Kassiotis; Kim J. Hasenkrug; Ulf Dittmer

It is well established that CD4+ T cells play an important role in immunity to infections with retroviruses such as HIV. However, in recent years CD4+ T cells have been subdivided into several distinct populations that are differentially regulated and perform widely varying functions. Thus, it is important to delineate the separate roles of these subsets, which range from direct antiviral activities to potent immunosuppression. In this review, we discuss contributions from the major CD4+ T cell subpopulations to retroviral immunity. Fundamental concepts obtained from studies on numerous viral infections are presented along with a more detailed analysis of studies on murine Friend virus. The relevance of these studies to HIV immunology and immunotherapy is reviewed.


Journal of Virology | 2012

Codelivery of the Chemokine CCL3 by an Adenovirus-Based Vaccine Improves Protection from Retrovirus Infection

Ruth Lietz; Wibke Bayer; Teona Ontikatze; Lena Johrden; Matthias Tenbusch; Michael Storcksdieck genannt Bonsmann; Klaus Überla; Ulf Dittmer; Oliver Wildner

ABSTRACT Processing and presentation of vaccine antigens by professional antigen-presenting cells (APCs) is of great importance for the efficient induction of protective immunity. We analyzed whether the efficacy of an adenovirus-based retroviral vaccine can be enhanced by coadministration of adenovirus-encoded chemokines that attract and stimulate APCs. In the Friend retrovirus (FV) mouse model we coexpressed CCL3, CCL20, CCL21, or CXCL14 from adenoviral vectors, together with FV Gag and Env antigens, and then analyzed immune responses and protection from pathogenic FV infection. Although most tested chemokines did not improve protection against FV challenge, mice that received adenoviral vectors encoding CCL3 together with FV antigens showed significantly better control over viral loads and FV-induced disease than mice immunized with the viral antigens only. Improved protection correlated with enhanced virus-specific CD4+ T cell responses and higher neutralizing antibody titers. To apply these results to an HIV vaccine, mice were immunized with adenoviral vectors encoding the HIV antigens Env and Gag-Pol and coadministered vectors encoding CCL3. Again, this combination vaccine induced higher virus-specific antibody titers and CD4+ T cell responses than did the HIV antigens alone. These results indicate that coexpression of the chemokine CCL3 by adenovirus-based vectors may be a promising tool to improve antiretroviral vaccination strategies.


Scientific Reports | 2016

Broad and potent antiviral activity of the NAE inhibitor MLN4924.

Vu Thuy Khanh Le-Trilling; Dominik A. Megger; Benjamin Katschinski; Christine D. Landsberg; Meike U. Rückborn; Sha Tao; Adalbert Krawczyk; Wibke Bayer; Ingo Drexler; Matthias Tenbusch; Barbara Sitek; Mirko Trilling

In terms of infected human individuals, herpesviruses range among the most successful virus families. Subclinical herpesviral infections in healthy individuals contrast with life-threatening syndromes under immunocompromising and immunoimmature conditions. Based on our finding that cytomegaloviruses interact with Cullin Roc ubiquitin ligases (CRLs) in the context of interferon antagonism, we systematically assessed viral dependency on CRLs by utilizing the drug MLN4924. CRL activity is regulated through the conjugation of Cullins with the ubiquitin-like molecule Nedd8. By inhibiting the Nedd8-activating Enzyme (NAE), MLN4924 interferes with Nedd8 conjugation and CRL activity. MLN4924 exhibited pronounced antiviral activity against mouse and human cytomegalovirus, herpes simplex virus (HSV)- 1 (including multi-drug resistant clinical isolates), HSV-2, adeno and influenza viruses. Human cytomegalovirus genome amplification was blocked at nanomolar MLN4924 concentrations. Global proteome analyses revealed that MLN4924 blocks cytomegaloviral replication despite increased IE1 amounts. Expression of dominant negative Cullins assigned this IE regulation to defined Cullin molecules and phenocopied the antiviral effect of MLN4924.


Cell Reports | 2016

Opposing Development of Cytotoxic and Follicular Helper CD4 T Cells Controlled by the TCF-1-Bcl6 Nexus

Tiziano Donnarumma; George R. Young; Julia Merkenschlager; Urszula Eksmond; Nadine Bongard; Stephen L. Nutt; Claude Boyer; Ulf Dittmer; Vu Thuy Khanh Le-Trilling; Mirko Trilling; Wibke Bayer; George Kassiotis

Summary CD4+ T cells develop distinct and often contrasting helper, regulatory, or cytotoxic activities. Typically a property of CD8+ T cells, granzyme-mediated cytotoxic T cell (CTL) potential is also exerted by CD4+ T cells. However, the conditions that induce CD4+ CTLs are not entirely understood. Using single-cell transcriptional profiling, we uncover a unique signature of Granzyme B (GzmB)+ CD4+ CTLs, which distinguishes them from other CD4+ T helper (Th) cells, including Th1 cells, and strongly contrasts with the follicular helper T (Tfh) cell signature. The balance between CD4+ CTL and Tfh differentiation heavily depends on the class of infecting virus and is jointly regulated by the Tfh-related transcription factors Bcl6 and Tcf7 (encoding TCF-1) and by the expression of the inhibitory receptors PD-1 and LAG3. This unique profile of CD4+ CTLs offers targets for their study, and its antagonism by the Tfh program separates CD4+ T cells with either helper or killer functions.


Virology Journal | 2013

Comparison of polystyrene nanoparticles and UV-inactivated antigen-displaying adenovirus for vaccine delivery in mice

Lena Johrden; Matthias Tenbusch; Ruth Lietz; Michael Storcksdieck genannt Bonsmann; Thomas Niezold; Oliver Wildner; Wibke Bayer

BackgroundInert nanoparticles are attracting attention as carriers for protein-based vaccines. Here we evaluate the immunogenicity of the model antigen ovalbumin delivered on polystyrene particles and directly compare particulate delivery with adenovirus-based immunization.FindingsMice were vaccinated with soluble ovalbumin, ovalbumin-coated polystyrene particles of different sizes, or an adenovirus-based expression-display vector that encodes and displays a pIX-ovalbumin fusion protein. Antibody responses were clearly higher when ovalbumin was administered on polystyrene particles compared to soluble protein administration, regardless of the particle size. Compared to adenovirus-based immunization, antibody levels were lower if an equivalent amount of protein was delivered, and no cellular immune response was detectable.ConclusionsWe demonstrate in a side-by-side comparison that inert nanoparticles allow for the reduction of the administered antigen amount compared to immunization with soluble protein and induce strongly enhanced antibody responses, but responses are lower compared to adenovirus-based immunization.


Retrovirology | 2016

Combination of nanoparticle-based therapeutic vaccination and transient ablation of regulatory T cells enhances anti-viral immunity during chronic retroviral infection.

Torben Knuschke; Olga Rotan; Wibke Bayer; Viktoriya Sokolova; Wiebke Hansen; Tim Sparwasser; Ulf Dittmer; Matthias Epple; Jan Buer; Astrid M. Westendorf

BackgroundRegulatory T cells (Tregs) have been shown to limit anti-viral immunity during chronic retroviral infection and to restrict vaccine-induced T cell responses. The objective of the study was to assess whether a combinational therapy of nanoparticle-based therapeutic vaccination and concomitant transient ablation of Tregs augments anti-viral immunity and improves virus control in chronically retrovirus-infected mice. Therefore, chronically Friend retrovirus (FV)-infected mice were immunized with calcium phosphate (CaP) nanoparticles functionalized with TLR9 ligand CpG and CD8+ or CD4+ T cell epitope peptides (GagL85–93 or Env gp70123–141) of FV. In addition, Tregs were ablated during the immunization process. Reactivation of CD4+ and CD8+ effector T cells was analysed and the viral loads were determined.ResultsTherapeutic vaccination of chronically FV-infected mice with functionalized CaP nanoparticles transiently reactivated cytotoxic CD8+ T cells and significantly reduced the viral loads. Transient ablation of Tregs during nanoparticle-based therapeutic vaccination strongly enhanced anti-viral immunity and further decreased viral burden.ConclusionOur data illustrate a crucial role for CD4+ Foxp3+ Tregs in the suppression of anti-viral T cell responses during therapeutic vaccination against chronic retroviral infection. Thus, the combination of transient Treg ablation and therapeutic nanoparticle-based vaccination confers robust and sustained anti-viral immunity.

Collaboration


Dive into the Wibke Bayer's collaboration.

Top Co-Authors

Avatar

Ulf Dittmer

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sonja Windmann

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Astrid M. Westendorf

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ruth Lietz

Ruhr University Bochum

View shared research outputs
Top Co-Authors

Avatar

Jan Buer

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge