Network


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

Hotspot


Dive into the research topics where Ans De Beuckelaer is active.

Publication


Featured researches published by Ans De Beuckelaer.


Proceedings of the National Academy of Sciences of the United States of America | 2016

PH-degradable imidazoquinoline-ligated nanogels for lymph node-focused immune activation

Lutz Nuhn; Nane Vanparijs; Ans De Beuckelaer; Lien Lybaert; G. Verstraete; Kim Deswarte; Stefan Lienenklaus; Nikunj M. Shukla; Alex C. D. Salyer; Bart N. Lambrecht; Johan Grooten; Sunil A. David; Stefaan De Koker; Bruno G. De Geest

Significance The newest generation of small-molecule vaccine adjuvants aims at triggering specific receptors expressed by dendritic cells, the working horses of our immune system. Unfortunately, owing to their small size, upon administration these molecules rapidly enter systemic circulation and cause systemic inflammation. We report on a nanotechnology-based solution for this issue by covalent ligation of a potent immunostimulatory small molecule to hydrogel nanoparticles. This approach allows for lymph node-restricted immune activation and avoids systemic dissemination. Importantly, relative to soluble immunostimulatory compound, nanoparticle ligation yields increased immune activation in the draining lymph nodes and results in strongly increased antibody titers and T-cell responses against an admixed vaccine antigen. Agonists of Toll-like receptors (TLRs) are potent activators of the innate immune system and hold promise as vaccine adjuvant and for anticancer immunotherapy. Unfortunately, in soluble form they readily enter systemic circulation and cause systemic inflammatory toxicity. Here we demonstrate that by covalent ligation of a small-molecule imidazoquinoline-based TLR7/8 agonist to 50-nm-sized degradable polymeric nanogels the potency of the agonist to activate TLR7/8 in in vitro cultured dendritic cells is largely retained. Importantly, imidazoquinoline-ligated nanogels focused the in vivo immune activation on the draining lymph nodes while dramatically reducing systemic inflammation. Mechanistic studies revealed a prevalent passive diffusion of the nanogels to the draining lymph node. Moreover, immunization studies in mice have shown that relative to soluble TLR7/8 agonist, imidazoquinoline-ligated nanogels induce superior antibody and T-cell responses against a tuberculosis antigen. This approach opens possibilities to enhance the therapeutic benefit of small-molecule TLR agonist for a variety of applications.


ACS Applied Materials & Interfaces | 2016

Spontaneous Protein Adsorption on Graphene Oxide Nanosheets Allowing Efficient Intracellular Vaccine Protein Delivery

Hui Li; Kaat Fierens; Zhiyue Zhang; Nane Vanparijs; Martijn J. Schuijs; Katleen Van Steendam; Natàlia Feiner Gracia; Riet De Rycke; Thomas De Beer; Ans De Beuckelaer; Stefaan De Koker; Dieter Deforce; Lorenzo Albertazzi; Johan Grooten; Bart N. Lambrecht; Bruno G. De Geest

Nanomaterials hold potential of altering the interaction between therapeutic molecules and target cells or tissues. High aspect ratio nanomaterials in particular have been reported to possess unprecedented properties and are intensively investigated for their interaction with biological systems. Graphene oxide (GOx) is a water-soluble graphene derivative that combines high aspect ratio dimension with functional groups that can be exploited for bioconjugation. Here, we demonstrate that GOx nanosheets can spontaneously adsorb proteins by a combination of interactions. This property is then explored for intracellular protein vaccine delivery, in view of the potential of GOx nanosheets to destabilize lipid membranes such as those of intracellular vesicles. Using a series of in vitro experiments, we show that GOx nanosheet adsorbed proteins are efficiently internalized by dendritic cells (DCs: the most potent class of antigen presenting cells of the immune system) and promote antigen cross-presentation to CD8 T cells. The latter is a hallmark in the induction of potent cellular antigen-specific immune responses against intracellular pathogens and cancer.


Vaccine | 2015

The site of administration influences both the type and the magnitude of the immune response induced by DNA vaccine electroporation

Gaëlle Vandermeulen; Kevin Vanvarenberg; Ans De Beuckelaer; Stefaan De Koker; Laure Lambricht; Catherine Uyttenhove; Anca Reschner; Alain Vanderplasschen; Johan Grooten; Véronique Préat

We investigated the influence of the site of administration of DNA vaccine on the induced immune response. DNA vaccines were administered by electroporation at three different sites: tibial cranial muscle, abdominal skin and ear pinna. Aiming to draw general conclusions about DNA vaccine delivery, we successively used several plasmids encoding either luciferase and ovalbumin as models or gp160 and P1A as vaccines against HIV and P815 mastocytoma, respectively. Low levels and duration of luciferase transgene expression were observed after electroporation of the abdominal skin, partly explaining its lower immunogenic performance as compared to the other sites of administration. Analyses of OT-I CD8+ and OT-II CD4+ T cell responses highlighted the differential impact of the delivery site on the elicited immune response. Muscle electroporation induced the strongest humoral immune response and both muscle and ear pinna sites induced cellular immunity against gp160. Ear pinna delivery generated the highest level of CTL responses against P1A but electroporation of muscle and ear pinna were equally efficient in delaying P815 growth and improving mice survival. The present study demonstrated that the site of administration is a key factor to be tested in the development of DNA vaccine.


Advanced Healthcare Materials | 2017

Arginine-Rich Peptide-Based mRNA Nanocomplexes Efficiently Instigate Cytotoxic T Cell Immunity Dependent on the Amphipathic Organization of the Peptide.

Vimal Kumar Udhayakumar; Ans De Beuckelaer; Joanne McCaffrey; Cian M. McCrudden; Jonathan L. Kirschman; Daryll Vanover; Lien Van Hoecke; Kenny Roose; Kim Deswarte; Bruno G. De Geest; Stefan Lienenklaus; Philip J. Santangelo; Johan Grooten; Helen O. McCarthy; Stefaan De Koker

To date, the mRNA delivery field has been heavily dominated by lipid-based systems. Reports on the use of nonlipid carriers for mRNA delivery in contrast are rare in the context of mRNA vaccination. This paper describes the potential of a cell-penetrating peptide containing the amphipathic RALA motif to deliver antigen-encoding mRNA to the immune system. RALA condenses mRNA into nanocomplexes that display acidic pH-dependent membrane disruptive properties. RALA mRNA nanocomplexes enable mRNA escape from endosomes and thereby allow expression of mRNA inside the dendritic cell cytosol. Strikingly, RALA mRNA nanocomplexes containing pseudouridine and 5-methylcytidine modified mRNA elicit potent cytolytic T cell responses against the antigenic mRNA cargo and show superior efficacy in doing so when compared to RALA mRNA nanocomplexes containing unmodified mRNA. RALAs unique sequence and structural organization are vital to act as mRNA vaccine vehicle, as arginine-rich peptide variants that lack the RALA motif show reduced mRNA complexation, impaired cellular uptake and lose the ability to transfect dendritic cells in vitro and to evoke T cell immunity in vivo.


Trends in Molecular Medicine | 2017

Type I Interferons Modulate CD8+ T Cell Immunity to mRNA Vaccines

Ans De Beuckelaer; Johan Grooten; Stefaan De Koker

mRNA vaccines have emerged as potent tools to elicit antitumor T cell immunity. They are characterized by a strong induction of type I interferons (IFNs), potent inflammatory cytokines affecting T cell differentiation and survival. Recent reports have attributed opposing roles for type I IFNs in modulating CD8+ T cell immunity to mRNA vaccines, from profoundly stimulatory to strongly inhibitory. The mechanisms behind this duality are unclear. Disentangling the factors governing the beneficial or detrimental impact of type I IFNs on CD8+ T cell responses is vital to the design of mRNA vaccines of increased potency. In light of recent advancements regarding the complex role of type I IFNs in regulating CD8+ T cell immunity to infectious diseases, we posit that the dual outcome of type I IFNs on CD8+ T cell responses to mRNA vaccination is determined by the timing and intensity of type I IFN induction relative to T cell receptor (TCR) activation.


Molecular Therapy | 2016

Type I Interferons Interfere with the Capacity of mRNA Lipoplex Vaccines to Elicit Cytolytic T Cell Responses

Ans De Beuckelaer; Charlotte Pollard; Sandra Van Lint; Kenny Roose; Lien Van Hoecke; Thomas Naessens; Vimal Kumar Udhayakumar; Muriel Smet; Niek N. Sanders; Stefan Lienenklaus; Xavier Saelens; Siegfried Weiss; Guido Vanham; Johan Grooten; Stefaan De Koker

Given their high potential to evoke cytolytic T cell responses, tumor antigen-encoding messenger RNA (mRNA) vaccines are now being intensively explored as therapeutic cancer vaccines. mRNA vaccines clearly benefit from wrapping the mRNA into nano-sized carriers such as lipoplexes that protect the mRNA from degradation and increase its uptake by dendritic cells in vivo. Nevertheless, the early innate host factors that regulate the induction of cytolytic T cells to mRNA lipoplex vaccines have remained unresolved. Here, we demonstrate that mRNA lipoplexes induce a potent type I interferon (IFN) response upon subcutaneous, intradermal and intranodal injection. Regardless of the route of immunization applied, these type I IFNs interfered with the generation of potent cytolytic T cell responses. Most importantly, blocking type I IFN signaling at the site of immunization through the use of an IFNAR blocking antibody greatly enhanced the prophylactic and therapeutic antitumor efficacy of mRNA lipoplexes in the highly aggressive B16 melanoma model. As type I IFN induction appears to be inherent to the mRNA itself rather than to unique properties of the mRNA lipoplex formulation, preventing type I IFN induction and/or IFNAR signaling at the site of immunization might constitute a widely applicable strategy to improve the potency of mRNA vaccination.


Immunity, inflammation and disease | 2016

Cholesterol‐sensing liver X receptors stimulate Th2‐driven allergic eosinophilic asthma in mice

Muriel Smet; Lien Van Hoecke; Ans De Beuckelaer; Seppe Vander Beken; Thomas Naessens; Karl Vergote; Monique Willart; Bart N. Lambrecht; Jan Åke Gustafsson; Knut R. Steffensen; Johan Grooten

Liver X receptors (LXRs) are nuclear receptors that function as cholesterol sensors and regulate cholesterol homeostasis. High cholesterol has been recognized as a risk factor in asthma; however, the mechanism of this linkage is not known.


European Journal of Immunology | 2016

Mycobacterium tuberculosis-associated synthetic mycolates differentially exert immune stimulatory adjuvant activity

Muriel Smet; Charlotte Pollard; Ans De Beuckelaer; Lien Van Hoecke; Seppe Vander Beken; Stefaan De Koker; Juma'a R. Al Dulayymi; Kris Huygen; Jan A. Verschoor; Mark S. Baird; Johan Grooten

Mycolic acids (MAs) are highly hydrophobic long‐chain α‐alkyl β‐hydroxy fatty acids present in the cell wall of Mycobacterium tuberculosis (Mtb) as a complex mixture of molecules with a common general structure but with variable functional groups in the meromycolate chain. In this study, we addressed the relationship between the MA molecular structure and their contribution to the development of T‐cell immune responses. Hereto, we used the model antigen ovalbumin and single synthetic MAs, differing in oxygenation class and cis versus trans proximal cyclopropane configuration, as immune stimulatory agents. Subcutaneous delivery of liposome‐formulated MAs with a proximal cis cyclopropane elicited antigen‐specific Th1 and cytotoxic T‐cell immune responses, whereas intratracheal immunization elicited pulmonary Th17 responses. These immune stimulatory activities depended not only on the cis versus trans proximal cyclopropane configuration but also on the MA oxygenation class. Our study thus shows that both the presence and nature of the functional groups in the meromycolate chain affect the immune stimulatory adjuvant activity of Mtb mycolates and suggests that Mtb bacilli may impact on the host protective immune response by modulating the cis versus trans stereochemistry of its mycolates as well as by altering the oxygenation class of the meromycolate functional group.


Molecular Therapy | 2016

Coadministration of a Plasmid Encoding HIV-1 Gag Enhances the Efficacy of Cancer DNA Vaccines

Laure Lambricht; Kevin Vanvarenberg; Ans De Beuckelaer; Lien Van Hoecke; Johan Grooten; Bernard Ucakar; Pascale Lipnik; Niek N. Sanders; Stefan Lienenklaus; Véronique Préat; Gaëlle Vandermeulen

DNA vaccination holds great promise for the prevention and treatment of cancer and infectious diseases. However, the clinical ability of DNA vaccines is still controversial due to the limited immune response initially observed in humans. We hypothesized that electroporation of a plasmid encoding the HIV-1 Gag viral capsid protein would enhance cancer DNA vaccine potency. DNA electroporation used to deliver plasmids in vivo, induced type I interferons, thereby supporting the activation of innate immunity. The coadministration of ovalbumin (OVA) and HIV-1 Gag encoding plasmids modulated the adaptive immune response. This strategy favored antigen-specific Th1 immunity, delayed B16F10-OVA tumor growth and improved mouse survival in both prophylactic and therapeutic vaccination approaches. Similarly, a prophylactic DNA immunization against the melanoma-associated antigen gp100 was enhanced by the codelivery of the HIV-1 Gag plasmid. The adjuvant effect was not driven by the formation of HIV-1 Gag virus-like particles. This work highlights the ability of both electroporation and the HIV-1 Gag plasmid to stimulate innate immunity for enhancing cancer DNA vaccine immunogenicity and demonstrates interesting tracks for the design of new translational genetic adjuvants to overcome the current limitations of DNA vaccines in humans.


Scientific Reports | 2017

Inflammatory monocytes regulate Th1 oriented immunity to CpG adjuvanted protein vaccines through production of IL-12

Stefaan De Koker; Lien Van Hoecke; Ans De Beuckelaer; Kenny Roose; Kim Deswarte; Monique Willart; Pieter Bogaert; Thomas Naessens; Bruno G. De Geest; Xavier Saelens; Bart N. Lambrecht; Johan Grooten

Due to their capacity to skew T cell responses towards Th1 oriented immunity, oligonucleotides containing unmethylated CpG motifs (CpG) have emerged as interesting adjuvants for vaccination. Whereas the signalling pathways in response to CpG mediated TLR9 activation have been extensively documented at the level of the individual cell, little is however known on the precise identity of the innate immune cells that govern T cell priming and polarisation to CpG adjuvanted protein antigens in vivo. In this study, we demonstrate that optimal induction of Th1 oriented immunity to CpG adjuvanted protein vaccines requires the coordinated actions of conventional DCs and of monocytes. Whilst conventional DCs were required for antigen presentation and initial T cell priming, monocytes constitute the main source of the Th1 polarising cytokine IL-12.

Collaboration


Dive into the Ans De Beuckelaer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge