Network


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

Hotspot


Dive into the research topics where Ellen Van Gulck is active.

Publication


Featured researches published by Ellen Van Gulck.


Molecular Therapy | 2013

Type I IFN Counteracts the Induction of Antigen-Specific Immune Responses by Lipid-Based Delivery of mRNA Vaccines

Charlotte Pollard; Joanna Rejman; Winni De Haes; Bernard Verrier; Ellen Van Gulck; Thomas Naessens; Stefaan C. De Smedt; Pieter Bogaert; Johan Grooten; Guido Vanham; Stefaan De Koker

The use of DNA and viral vector-based vaccines for the induction of cellular immune responses is increasingly gaining interest. However, concerns have been raised regarding the safety of these immunization strategies. Due to the lack of their genome integration, mRNA-based vaccines have emerged as a promising alternative. In this study, we evaluated the potency of antigen-encoding mRNA complexed with the cationic lipid 1,2-dioleoyl-3trimethylammonium-propane/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOTAP/DOPE ) as a novel vaccination approach. We demonstrate that subcutaneous immunization of mice with mRNA encoding the HIV-1 antigen Gag complexed with DOTAP/DOPE elicits antigen-specific, functional T cell responses resulting in specific killing of Gag peptide-pulsed cells and the induction of humoral responses. In addition, we show that DOTAP/DOPE complexed antigen-encoding mRNA displays immune-activating properties characterized by secretion of type I interferon (IFN) and the recruitment of proinflammatory monocytes to the draining lymph nodes. Finally, we demonstrate that type I IFN inhibit the expression of DOTAP/DOPE complexed antigen-encoding mRNA and the subsequent induction of antigen-specific immune responses. These results are of high relevance as they will stimulate the design and development of improved mRNA-based vaccination approaches.


Biomaterials | 2012

Electrospun cellulose acetate phthalate fibers for semen induced anti-HIV vaginal drug delivery

Chaobo Huang; Stefaan Soenen; Ellen Van Gulck; Guido Vanham; Joanna Rejman; Serge Van Calenbergh; Chris Vervaet; Tom Coenye; Hans Verstraelen; Marleen Temmerman; Jo Demeester; Stefaan C. De Smedt

Despite many advances in modern medicine, human immunodeficiency virus (HIV) still affects the health of millions of people world-wide and much effort is put in developing methods to either prevent infection or to eradicate the virus after infection has occurred. Here, we describe the potential use of electrospun cellulose acetate phthalate (CAP) fibers as a tool to prevent HIV transmission. During the electrospinning process, anti-viral drugs can easily be incorporated in CAP fibers. Interestingly, as a result of the pH-dependent solubility of CAP, the fibers are stable in vaginal fluid (the healthy vaginal flora has a pH of below 4.5), whereas the addition of small amounts of human semen (pH between 7.4 and 8.4) immediately dissolves the fibers which results in the release of the encapsulated drugs. The pH-dependent release properties have been carefully studied and we show that the released anti-viral drugs, together with the CAP which has been reported to have intrinsic antimicrobial activity, efficiently neutralize HIV in vitro.


Journal of Immunotherapy | 2006

Simultaneous Activation of Viral Antigen-specific Memory Cd4+ and Cd8+ T-cells Using mrna-electroporated Cd40-activated Autologous B-cells

Glenn Van den Bosch; Ellen Van Gulck; Peter Ponsaerts; Griet Nijs; Marc Lenjou; Ludwig Apers; Ilse Kint; Leo Heyndrickx; Guido Vanham; Dirk R. Van Bockstaele; Zwi N. Berneman; Viggo Van Tendeloo

Recently, it has become obvious that not only CD8+ T-cells, but also CD4+ T-helper cells are required for the induction of an effective, long-lasting cellular immune response. In view of the clinical importance of cytomegalovirus (CMV) and human immunodeficiency virus (HIV) infection, we developed 2 strategies to simultaneously reactivate viral antigen-specific memory CD4+ and CD8+ T-cells of CMV-seropositive and HIV-seropositive subjects using mRNA-electroporated autologous CD40-activated B cells. In the setting of HIV, we provide evidence that CD40-activated B cells can be cultured from HAART-naive HIV-1 seropositive patients. These cells not only express and secrete the HIV p24 antigen after electroporation with codon-optimized HIV-1 gag mRNA, but can also be used to in vitro reactivate Gag antigen-specific interferon-γ–producing CD4+ and CD8+ autologous T-cells. For the CMV-specific approach, we applied mRNA coding for the pp65 protein coupled to the lysosomal-associated membrane protein-1 to transfect CD40-activated B cells to induce CMV antigen-specific CD4+ and CD8+ T-cells. More detailed analysis of the activated interferon-γ–producing CMV pp65 tetramer positive CD8+ T-cells revealed an effector memory phenotype with the capacity to produce interleukin-2. Our findings clearly show that the concomitant activation of both CD4+ and CD8+ (memory) T-cells using mRNA-electroporated CD40-B cells is feasible in CMV and HIV-1–seropositive persons, which indicates the potential value of this approach for application in cellular immunotherapy of infectious diseases.


PLOS ONE | 2012

An Improved Protocol for Efficient Engraftment in NOD/LTSZ-SCIDIL-2RγNULL Mice Allows HIV Replication and Development of Anti-HIV Immune Responses

Maneesh Singh; Pratibha Singh; Gilles Gaudray; Lucia Musumeci; Caroline Thielen; Dolores Vaira; Claire Vandergeeten; Laurence Delacroix; Ellen Van Gulck; Guido Vanham; Laurence de Leval; Souad Rahmouni; Michel Moutschen

Cord blood hematopoietic progenitor cells (CB-HPCs) transplanted immunodeficient NOD/LtsZ-scidIL2Rγnull (NSG) and NOD/SCID/IL2Rγnull (NOG) mice need efficient human cell engraftment for long-term HIV-1 replication studies. Total body irradiation (TBI) is a classical myeloablation regimen used to improve engraftment levels of human cells in these humanized mice. Some recent reports suggest the use of busulfan as a myeloablation regimen to transplant HPCs in neonatal and adult NSG mice. In the present study, we further ameliorated the busulfan myeloablation regimen with fresh CB-CD34+cell transplantation in 3–4 week old NSG mice. In this CB-CD34+transplanted NSG mice engraftment efficiency of human CD45+cell is over 90% in peripheral blood. Optimal engraftment promoted early and increased CD3+T cell levels, with better lymphoid tissue development and prolonged human cell chimerism over 300 days. These humanized NSG mice have shown long-lasting viremia after HIV-1JRCSF and HIV-1Bal inoculation through intravenous and rectal routes. We also saw a gradual decline of the CD4+T cell count, widespread immune activation, up-regulation of inflammation marker and microbial translocation after HIV-1 infection. Humanized NSG mice reconstituted according to our new protocol produced, moderate cellular and humoral immune responses to HIV-1 postinfection. We believe that NSG mice reconstituted according to our easy to use protocol will provide a better in vivo model for HIV-1 replication and anti-HIV-1 therapy trials.


PLOS ONE | 2012

Immune and Viral Correlates of "Secondary Viral Control" after Treatment Interruption in Chronically HIV-1 Infected Patients

Ellen Van Gulck; Lotte L. Bracke; Leo Heyndrickx; Sandra Coppens; Derek Atkinson; Céline Merlin; Alexander O. Pasternak; Eric Florence; Guido Vanham

Upon interruption of antiretroviral therapy, HIV-infected patients usually show viral load rebound to pre-treatment levels. Four patients, hereafter referred to as secondary controllers (SC), were identified who initiated therapy during chronic infection and, after stopping treatment, could control virus replication at undetectable levels for more than six months. In the present study we set out to unravel possible viral and immune parameters or mechanisms of this phenomenon by comparing secondary controllers with elite controllers and non-controllers, including patients under HAART. As candidate correlates of protection, virus growth kinetics, levels of intracellular viral markers, several aspects of HIV-specific CD4+ and CD8+ T cell function and HIV neutralizing antibodies were investigated. As expected all intracellular viral markers were lower in aviremic as compared to viremic subjects, but in addition both elite and secondary controllers had lower levels of viral unspliced RNA in PBMC as compared to patients on HAART. Ex vivo cultivation of the virus from CD4+ T cells of SC consistently failed in one patient and showed delayed kinetics in the three others. Formal in vitro replication studies of these three viruses showed low to absent growth in two cases and a virus with normal fitness in the third case. T cell responses toward HIV peptides, evaluated in IFN-γ ELISPOT, revealed no significant differences in breadth, magnitude or avidity between SC and all other patient groups. Neither was there a difference in polyfunctionality of CD4+ or CD8+ T cells, as evaluated with intracellular cytokine staining. However, secondary and elite controllers showed higher proliferative responses to Gag and Pol peptides. SC also showed the highest level of autologous neutralizing antibodies. These data suggest that higher T cell proliferative responses and lower replication kinetics might be instrumental in secondary viral control in the absence of treatment.


Molecular Therapy | 2010

Polyelectrolyte Capsules-containing HIV-1 p24 and Poly I:C Modulate Dendritic Cells to Stimulate HIV-1-specific Immune Responses

Winni De Haes; Stefaan De Koker; Charlotte Pollard; Derek Atkinson; Erika Vlieghe; Jessy Hoste; Joanna Rejman; Stefaan C. De Smedt; Johan Grooten; Guido Vanham; Ellen Van Gulck

Polyelectrolyte microcapsules (MCs) are potent protein delivery vehicles which can be tailored with ligands to stimulate maturation of dendritic cells (DCs). We investigated the immune stimulatory capacity of monocyte-derived DC (Mo-DC) loaded with these MCs, containing p24 antigen from human immunodeficiency virus type 1 (HIV-1) alone [p24-containing MC (MCp24)] or with the Toll-like receptor ligand 3 (TLR3) ligand poly I:C (MCp24pIC) as a maturation factor. MO-DC, loaded with MCp24pIC, upregulated CCR7, CD80, CD83, and CD86 and produced high amounts of interleukin-12 (IL-12) cytokine, to a similar extent as MCp24 in the presence of an optimized cytokine cocktail. MO-DC from HIV-infected patients under highly active antiretroviral therapy (HAART) exposed to MCp24 together with cytokine cocktail or to MCp24pIC expanded autologous p24-specific CD4(+) and CD8(+) T-cell responses as measured by interferon-gamma (IFN-gamma) and IL-2 cytokine production and secretion. In vivo relevance was shown by immunizing C57BL/6 mice with MCp24pIC, which induced both humoral and cellular p24-specific immune responses. Together these data provide a proof of principle that both antigen and DC maturation signal can be delivered as a complex with polyelectrolyte capsules to stimulate virus-specific T cells both in vitro and in vivo. Polyelectrolyte MCs could be useful for in vivo immunization in HIV-1 and other infections.


Journal of Virology | 2013

Targeting of human antigen-presenting cell subsets

Cleo Goyvaerts; Jozef Dingemans; Kurt De Groeve; Carlo Heirman; Ellen Van Gulck; Guido Vanham; Patrick De Baetselier; Kris Thielemans; Geert Raes; Karine Breckpot

ABSTRACT Antigen-presenting cells are a heterogeneous group of cells that are characterized by their functional specialization. Consequently, targeting specific antigen-presenting cell subsets offers opportunities to induce distinct T cell responses. Here we report on the generation and use of nanobodies (Nbs) to target lentivectors specifically to human lymph node-resident myeloid dendritic cells, demonstrating that Nbs represent a powerful tool to redirect lentivectors to human antigen-presenting cell subsets.


European Journal of Immunology | 2012

CD34-derived dendritic cells transfected ex vivo with HIV-Gag mRNA induce polyfunctional T-cell responses in nonhuman primates.

Gabrielle Romain; Ellen Van Gulck; Olivier Epaulard; SangKon Oh; Dapeng Li; Gerard Zurawski; Sandra Zurawski; Antonio Cosma; Lucille Adam; Catherine Chapon; Biliana Todorova; Jacques Banchereau; Nathalie Dereuddre-Bosquet; Guido Vanham; Roger Le Grand; Frédéric Martinon

The pivotal role of DCs in initiating immune responses led to their use as vaccine vectors. However, the relationship between DC subsets involved in antigen presentation and the type of elicited immune responses underlined the need for the characterization of the DCs generated in vitro. The phenotypes of tissue‐derived APCs from a cynomolgus macaque model for human vaccine development were compared with ex vivo‐derived DCs. Monocyte/macrophages predominated in bone marrow (BM) and blood. Myeloid DCs (mDCs) were present in all tested tissues and were more highly represented than plasmacytoid DCs (pDCs). As in human skin, Langerhans cells (LCs) resided exclusively in the macaque epidermis, expressing CD11c, high levels of CD1a and langerin (CD207). Most DC subsets were endowed with tissue‐specific combinations of PRRs. DCs generated from CD34+ BM cells (CD34‐DCs) were heterogeneous in phenotype. CD34‐DCs shared properties (differentiation and PRR) of dermal and epidermal DCs. After injection into macaques, CD34‐DCs expressing HIV‐Gag induced Gag‐specific CD4+ and CD8+ T cells producing IFN‐γ, TNF‐α, MIP‐1β, or IL‐2. In high responding animals, the numbers of polyfunctional CD8+ T cells increased with the number of booster injections. This DC‐based vaccine strategy elicited immune responses relevant to the DC subsets generated in vitro.


Current HIV Research | 2010

Role of dendritic cells in HIV-immunotherapy.

Ellen Van Gulck; Viggo Van Tendeloo; Zwi N. Berneman; Guido Vanham

HIV remains one of the most important deadly infections today, due to the lack of a preventive vaccine and limited access to medical care in developing countries. In developed countries, antiretroviral therapy is available, but it can not eliminate the virus, implying that life-long therapy is necessary. Therefore, it is important that other strategies such as therapeutic vaccination will be developed to control virus replication or even eliminate the virus. The major obstacles towards such a strategy are the huge variability of the virus and the profound HIV-induced immune dysfunction. In this review we focus on dendritic cell based immunotherapies against HIV. To develop an efficient immunotherapy, several elements should be taken into account such as which antigen and loading strategy to use, how to deliver the immunogen, how to optimize the interaction between antigenic peptide and T cells and avoid tolerance. Clearly, to develop an immunotherapy to complement the effect of HAART, it is not sufficient to enhance T cell responses against a consensus sequence or against the prevailing plasma virus. Broad and potent immune response are needed to suppress the entire quasispecies, including the latent reservoir, and to prevent any escape.


Nanomedicine: Nanotechnology, Biology and Medicine | 2013

Lipoplexes carrying mRNA encoding Gag protein modulate dendritic cells to stimulate HIV-specific immune responses

Winni De Haes; Joanna Rejman; Charlotte Pollard; Céline Merlin; Marc Vekemans; Eric Florence; Stefaan C. De Smedt; Johan Grooten; Guido Vanham; Stefaan De Koker; Ellen Van Gulck

AIM Cationic lipids (Lipofectamine™ [Invitrogen, Merelbeke, Belgium] and 1,2-dioleoyl-3-trimethylammonium-propane/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine) and polymers (jetPEI™ and in vivo-jetPEI™ [Polyplus-transfection, Illkirch, France]) were evaluated for their potential to deliver mRNA to monocyte-derived dendritic cells. MATERIALS & METHODS Lipoplexes and polyplexes, containing mRNA encoding GFP or Gag protein, were incubated with human monocyte-derived dendritic cells and transfection efficiencies were assessed by flow cytometry. RESULTS Lipofectamine was by far the most efficient in mRNA delivery, therefore it was used in further experiments. Incubation of monocyte-derived dendritic cells isolated from HIV-1-positive donors with mRNA encoding Gag protein complexed to Lipofectamine resulted in 50% transfection. Importantly, coculture of these Gag-transfected dendritic cells with autologous T cells induced an over tenfold expansion of IFN-γ- and IL-2-secreting CD4(+) and CD8(+) T cells. CONCLUSION Cationic lipid-mediated mRNA delivery may be a useful tool for therapeutic vaccination against HIV-1. This approach can be applied to develop vaccination strategies for other infectious diseases and cancer.

Collaboration


Dive into the Ellen Van Gulck's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leo Heyndrickx

Institute of Tropical Medicine Antwerp

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Winni De Haes

Institute of Tropical Medicine Antwerp

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Derek Atkinson

Institute of Tropical Medicine Antwerp

View shared research outputs
Researchain Logo
Decentralizing Knowledge