Network


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

Hotspot


Dive into the research topics where Hakan Kalay is active.

Publication


Featured researches published by Hakan Kalay.


Molecular Immunology | 2009

Targeting glycan modified OVA to murine DC-SIGN transgenic dendritic cells enhances MHC class I and II presentation

Satwinder Kaur Singh; Johannes Stephani; Martin Schaefer; Hakan Kalay; Juan J. Garcia-Vallejo; Joke M. M. den Haan; Eirikur Saeland; Tim Sparwasser; Yvette van Kooyk

Dendritic cells have gained much interest in the field of anti-cancer vaccine development because of their central function in immune regulation. One of the receptors that facilitate DC-specific targeting of antigens is the DC-specific C-type lectin DC-SIGN. Although DC-SIGN is specifically expressed on human DCs, its murine homologue is not present on any murine DC subsets, which makes in vivo evaluation of potential DC-SIGN targeting vaccines very difficult. Here we describe the use of DC-SIGN transgenic mice, as a good model system to evaluate DC-SIGN targeting vaccines. We demonstrate that glycan modification of OVA with DC-SIGN targeting glycans, targets antigen specifically to bone marrow (BM)** derived DCs and splenic DCs. Glycan modification of OVA with Lewis X or Lewis B oligosaccharides, that target DC-SIGN transgenic DCs, resulted in efficient 10-fold induction of OT-II compared to unmodified OVA. Interestingly, glycan modified OVA proteins were significantly cross-presented to OT-I T cells by wild type DC, 10-fold more than native OVA, and the expression of DC-SIGN further enhanced this cross-presentation. Targeting of glycosylated OVA was neither accompanied with any DC maturation, nor the production of inflammatory or anti-inflammatory cytokines. Thus, we conclude that glycan modification of antigens and targeting to DC-SIGN enhance both CD4 and CD8 T cell responses. Furthermore, our data demonstrate that DC-SIGN transgenic mice are valuable tool for optimisation and efficiency testing of DC vaccination strategies that are designed to target in particular the human DC-SIGN receptor.


Journal of Controlled Release | 2012

Glycan-modified liposomes boost CD4 + and CD8 + T-cell responses by targeting DC-SIGN on dendritic cells

Wendy W. J. Unger; Astrid J. van Beelen; Sven C. M. Bruijns; Medha Joshi; Cynthia M. Fehres; Louis van Bloois; Marleen I. Verstege; Martino Ambrosini; Hakan Kalay; Kamran Nazmi; Jan G. M. Bolscher; Erik Hooijberg; Tanja D. de Gruijl; Gert Storm; Yvette van Kooyk

Cancer immunotherapy requires potent tumor-specific CD8(+) and CD4(+) T-cell responses, initiated by dendritic cells (DCs). Tumor antigens can be specifically targeted to DCs in vivo by exploiting their expression of C-type lectin receptors (CLR), which bind carbohydrate structures on antigens, resulting in internalization and antigen presentation to T-cells. We explored the potential of glycan-modified liposomes to target antigens to DCs to boost murine and human T-cell responses. Since DC-SIGN is a CLR expressed on DCs, liposomes were modified with DC-SIGN-binding glycans Lewis (Le)(B) or Le(X). Glycan modification of liposomes resulted in increased binding and internalization by BMDCs expressing human DC-SIGN. In the presence of LPS, this led to 100-fold more efficient presentation of the encapsulated antigens to CD4(+) and CD8(+) T-cells compared to unmodified liposomes or soluble antigen. Similarly, incubation of human moDC with melanoma antigen MART-1-encapsulated liposomes coated with Le(X) in the presence of LPS led to enhanced antigen-presentation to MART-1-specific CD8(+) T-cell clones. Moreover, this formulation drove primary CD8(+) T-cells to differentiate into high numbers of tetramer-specific, IFN-γ-producing effector T-cells. Together, our data demonstrate the potency of a glycoliposome-based vaccine targeting DC-SIGN for CD4(+) and CD8(+) effector T-cell activation. This approach may offer improved options for treatment of cancer patients and opens the way to in situ DC-targeted vaccination.


Molecular Immunology | 2009

MUC1 in human milk blocks transmission of human immunodeficiency virus from dendritic cells to T cells

Eirikur Saeland; Marein A.W.P. de Jong; Alexey A. Nabatov; Hakan Kalay; Teunis B. H. Geijtenbeek; Yvette van Kooyk

Mother-to-child transmission of human immunodeficiency virus-1 (HIV-1) occurs frequently via breast-feeding. HIV-1 targets DC-SIGN+ dendritic cells (DCs) in mucosal areas that allow efficient transmission of the virus to T cells. Here, we demonstrate that the epithelial mucin MUC1, abundant in milk, efficiently bound to DC-SIGN on DC. The O-linked glycans within the mucin domain contained Lewis X structures, that were specifically recognized by the receptor. Interestingly, MUC1 prevented DC-SIGN-mediated transmission of HIV-1 from DCs to CD4+ T cells. We hypothesize that repetitive units of Lewis X, within the mucin domain, play an important role in inhibiting transmission of HIV-1 from mother to child.


Molecular Immunology | 2013

Glycan-based DC-SIGN targeting vaccines to enhance antigen cross-presentation

Yvette van Kooyk; Wendy W. J. Unger; Cynthia M. Fehres; Hakan Kalay; Juan J. Garcia-Vallejo

Dendritic cells are the most efficient professional antigen-presenting cells in pathogen recognition and play a pivotal role in the control of the immune response. Pathogen recognition is ensured by the expression of a vast variety of pattern-recognition receptors. Amongst them are C-type lectins, a large family of receptors characterized by a domain that - in many cases - mediates calcium-dependent glycan binding. C-type lectins facilitate antigen uptake for efficient processing and presentation and, in some cases, also trigger signaling to modulate T cell responses. These properties make C-type lectin receptors ideal candidates for the targeting of antigens to dendritic cells for vaccination. DC-SIGN is a paradigmatic example of C-type lectin receptors on dendritic cells that facilitate vaccination strategies. DC-SIGN is highly expressed on immature conventional dendritic cells, particularly at the mucosa and the dermis, where DCs first encounter pathogens, but also can easily be accessed for vaccination. Upon ligand binding, DC-SIGN rapidly internalizes and directs its cargo into the endo-lysosomal pathway, which results in MHC-II presentation. But antigens targeted to DC-SIGN are also presented efficiently to CD8(+) T cells, suggesting there is an additional endocytic route that leads to cross-presentation. Simultaneous triggering of DC-SIGN and TLRs results in the modulation of cytokine responses and facilitates cross-presentation to enhance CD4(+) and CD8(+) T cell responses. Because the glycan specificity of DC-SIGN has been characterized in detail, glycans can be used for the targeting of antigens to DCs in a DC-SIGN-dependent manner. Glycans represent a great advantage over monoclonal antibodies, they diminish the risk of side effects, are very small, and their production can rely entirely in organic chemistry approaches. Here, we discuss the capacity of glycan-based vaccines to enhance antigen-specific CD4(+) and CD8(+) T cell responses in human skin and mouse model systems.


Molecular Immunology | 2013

Multivalent glycopeptide dendrimers for the targeted delivery of antigens to dendritic cells

Juan J. Garcia-Vallejo; Martino Ambrosini; A. Overbeek; W.E. van Riel; Karien Bloem; W.W.J. Unger; Fabrizio Chiodo; Jan G. M. Bolscher; Kamran Nazmi; Hakan Kalay; Y. van Kooyk

Dendritic cells are the most powerful type of antigen presenting cells. Current immunotherapies targeting dendritic cells have shown a relative degree of success but still require further improvement. One of the most important issues to solve is the efficiency of antigen delivery to dendritic cells in order to achieve an appropriate uptake, processing, and presentation to Ag-specific T cells. C-type lectins have shown to be ideal receptors for the targeting of antigens to dendritic cells and allow the use of their natural ligands - glycans - instead of antibodies. Amongst them, dendritic cell-specific ICAM-3-grabbing non-integrin (DC-SIGN) is an interesting candidate due to its biological properties and the availability of its natural carbohydrate ligands. Using Le(b)-conjugated poly(amido amine) (PAMAM) dendrimers we aimed to characterize the optimal level of multivalency necessary to achieve the desired internalization, lysosomal delivery, Ag-specific T cell proliferation, and cytokine response. Increasing DC-SIGN ligand multivalency directly translated in an enhanced binding, which might also be interesting for blocking purposes. Internalization, routing to lysosomal compartments, antigen presentation and cytokine response could be optimally achieved with glycopeptide dendrimers carrying 16-32 glycan units. This report provides the basis for the design of efficient targeting of peptide antigens for the immunotherapy of cancer, autoimmunity and infectious diseases.


Journal of Experimental Medicine | 2014

CNS myelin induces regulatory functions of DC-SIGN-expressing, antigen-presenting cells via cognate interaction with MOG

Juan J. Garcia-Vallejo; Juan M. Ilarregui; Hakan Kalay; S. Chamorro; Nathalie Koning; W.W.J. Unger; Martino Ambrosini; V. Montserrat; Rosette J. Fernandes; Sven C. M. Bruijns; J.R.T. van Weering; N.J. Paauw; T. O’Toole; J. van Horssen; P. van der Valk; Kamran Nazmi; Jan G. M. Bolscher; Jeffrey J. Bajramovic; C. D. Dijkstra; Bert A. 't Hart; Y. van Kooyk

Human myelin oligodendrocyte glycoprotein is decorated with fucosylated N-glycans that are recognized by DC-SIGN+ DCs and microglia that control immune homeostasis.


European Journal of Immunology | 2011

Design of neo-glycoconjugates that target the mannose receptor and enhance TLR-independent cross-presentation and Th1 polarization.

Satwinder Kaur Singh; Ingeborg Streng-Ouwehand; Manja Litjens; Hakan Kalay; Sven Burgdorf; Eirikur Saeland; Christian Kurts; W.W.J. Unger; Yvette van Kooyk

Cross‐presentation is an important mechanism by which DCs present exogenous antigens on MHC‐I molecules, and activate CD8+ T cells, cells that are crucial for the elimination of tumors. We investigated the feasibility of exploiting the capacity of the mannose receptor (MR) to improve both cross‐presentation of tumor antigens and Th polarization, processes that are pivotal for the anti‐tumor potency of cytotoxic T cells. To this end, we selected two glycan ligands of the MR, 3‐sulfo‐LewisA and tri‐GlcNAc (N‐acetylglucosamine), to conjugate to the model antigen OVA and assessed in vitro the effect on antigen presentation and Th differentiation. Our results demonstrate that conjugation of either 3‐sulfo‐LewisA or tri‐GlcNAc specifically directs antigen to the MR. Both neo‐glycoconjugates showed, even at low doses, improved uptake as compared with native OVA, resulting in enhanced cross‐presentation. Using MR−/− and MyD88‐TRIFF−/− bone marrow‐derived DCs (BMDCs), we show that the cross‐presentation of the neo‐glycoconjugates is dependent on MR and independent of TLR‐mediated signaling. Whereas proliferation of antigen‐specific CD4+ T cells was unchanged, stimulation with neo‐glycoconjugate‐loaded DCs enhanced the generation of IFN‐γ‐producing T cells. We conclude that modification of antigen with either 3‐sulfo‐LewisA or tri‐GlcNAc enhances cross‐presentation and permits Th1 skewing, through specific targeting of the MR, which may be beneficial for DC‐based vaccination strategies to treat cancer.


Biological Chemistry | 2006

The human cathelicidin peptide LL-37 and truncated variants induce segregation of lipids and proteins in the plasma membrane of Candida albicans

Alice L. den Hertog; Jan van Marle; Enno C. I. Veerman; Marianne Valentijn-Benz; Kamran Nazmi; Hakan Kalay; Christian H. Grün; Wim van 't Hof; Jan G. M. Bolscher; Arie V. Nieuw Amerongen

Abstract The human cathelicidin peptide LL-37 and several truncated variants differ in their capability to transmigrate over the plasma membrane of Candida albicans. We investigated whether retention at the cell perimeter or membrane transmigration affects their membrane-disrupting activities and candidacidal properties. Using fluorescein-labeled peptides, we demonstrate that LL-37 and its C-terminally truncated peptide LL-31 remain permanently associated with the perimeter of the cell. The N-terminally truncated peptide RK-31 initially accumulated at the cell boundary, but transmigrated into the cytoplasm within 30 min. The C-terminally truncated peptide LL-25 transmigrated instantaneously into the cytoplasm. The ultrastructural effects on the plasma membrane were studied by freeze-fracture electron microscopy combined with filipin cytochemistry. All peptides, whether they transmigrated over the plasma membrane or not, induced phase separation in the plasma membrane. All peptides induced leakage of cell components, including nucleotides and proteins. Proteins were identified by SDS-PAGE in combination with mass spectrometry, which revealed that predominantly proteins smaller than 50 kDa had leaked out of C. albicans.


European Journal of Immunology | 2014

Topical rather than intradermal application of the TLR7 ligand imiquimod leads to human dermal dendritic cell maturation and CD8+ T-cell cross-priming.

Cynthia M. Fehres; Sven C. M. Bruijns; Astrid J. van Beelen; Hakan Kalay; Martino Ambrosini; Erik Hooijberg; Wendy W. J. Unger; Tanja D. de Gruijl; Yvette van Kooyk

Toll‐like receptor (TLR) ligands are attractive candidate adjuvants for therapeutic cancer vaccines, since TLR signaling stimulates and tunes both humoral and cellular immune responses induced by dendritic cells (DCs). Given that human skin contains a dense network of DCs, which are easily accessible via (intra‐)dermal delivery of vaccines, skin is actively explored as an antitumor vaccination site. Here we used a human skin explant model to explore the potential of TLR ligands as adjuvants for DC activation in their complex microenvironment. We show that topical application of Aldara skin cream, 5% of which comprises the TLR7 agonist imiquimod, significantly enhanced DC migration as compared with that resulting from intradermal injection of the TLR7/8 ligand R848 or the soluble form of imiquimod. Moreover, Aldara‐treated DCs showed highest levels of the costimulatory molecules CD86, CD83, CD40, and CD70. Topical Aldara induced the highest production of pro‐inflammatory cytokines in skin biopsies. When combined with intradermal peptide vaccination, Aldara‐stimulated DCs showed enhanced cross‐presentation of the melanoma antigen MART‐1, which resulted in increased priming and activation of MART‐1‐specific CD8+ T cells. These results point to advantageous effects of combining the topical application of Aldara with antitumor peptide vaccination.


International Immunology | 2013

Glycodendrimers prevent HIV transmission via DC-SIGN on dendritic cells

Juan J. Garcia-Vallejo; Nathalie Koning; Martino Ambrosini; Hakan Kalay; Ilona M. Vuist; Ramin Sarrami-Forooshani; Teunis B. H. Geijtenbeek; Yvette van Kooyk

Dendritic cells (DCs) are antigen-presenting cells efficient in capturing pathogens, and processing their antigenic determinants for presentation to antigen-specific T cells to induce robust immune responses. Their location at peripheral tissues and the expression of pattern-recognition receptors, among them DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), facilitates the capture of pathogens before spreading. However, some pathogens have developed strategies to escape the immune system. One of the most successful is HIV-1, which targets DC-SIGN for transport to the lymph node where the virus infects CD4(+) T cells. Contact of HIV-1 with DC-SIGN is thus the first event in the pathogenic cascade and, therefore, it is the primary target point for therapies aimed at HIV infection prevention. DC-SIGN recognizes specific glycans on HIV-1 and this interaction can be blocked by competitive inhibition through glycans. Although the affinity of glycans is relatively low, multivalency may increase avidity and the strength to compete with HIV-1 virions. We have designed multivalent dendrimeric compounds based on Lewis-type antigens that bind DC-SIGN with high selectivity and avidity and that effectively block gp120 binding to DC-SIGN and, consequently, HIV transmission to CD4(+) T cells. Binding to DC-SIGN and gp120 inhibition was higher on glycodendrimers with larger molecular diameter, indicating that the geometry of the compounds is an important factor determining their functionality. Our compounds elicited DC-SIGN internalization, a property of the receptor upon triggering, but did not affect the maturation status of DCs. Thus, Le(X) glycodendrimers could be incorporated into topic prophylactic approaches for the prevention of HIV-1 transmission.

Collaboration


Dive into the Hakan Kalay's collaboration.

Top Co-Authors

Avatar

Yvette van Kooyk

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martino Ambrosini

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sven C. M. Bruijns

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wendy W. J. Unger

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cynthia M. Fehres

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kamran Nazmi

Academic Center for Dentistry Amsterdam

View shared research outputs
Researchain Logo
Decentralizing Knowledge