Network


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

Hotspot


Dive into the research topics where Sandra Laban is active.

Publication


Featured researches published by Sandra Laban.


European Journal of Immunology | 2009

Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: differential role for PD-L1.

Wendy W. J. Unger; Sandra Laban; Fleur S. Kleijwegt; Arno R. van der Slik; Bart O. Roep

Specific therapy with modulated DC may restore immunological tolerance, thereby obviating the need for chronic immunosuppression in transplantation or autoimmunity. In this study we compared the tolerizing capacity of dexamethasone (Dex)‐ and 1α,25‐dihydroxyvitamin D3 (VD3)‐modulated DC. Treatment of monocytes with either VD3 or Dex resulted in DC with stable, semi‐mature phenotypes compared with standard DC, with intermediate levels of co‐stimulatory and MHC class II molecules, which remained unaltered after subsequent pro‐inflammatory stimulation. IL‐12p70 secretion was lost by VD3‐ and Dex‐DC, whereas IL‐10 secretion was unaffected. VD3‐DC distinctly produced large amounts of TNF‐α. Both VD3‐ and Dex‐DC possessed the capacity to convert CD4 T cells into IL‐10‐secreting Treg potently suppressing the proliferation of responder T cells. However, only Treg induced by VD3‐DC exhibited antigen specificity. VD3‐, but not Dex‐, DC expressed significant high levels of PD‐L1 (programmed death‐1 ligand), upon activation. Blockade of PD‐L1 during priming redirected T cells to produce IFN‐γ instead of IL‐10 and abolished acquisition of regulatory capacity. Our findings demonstrate that both VD3‐ and Dex‐DC possess durable but differential tolerogenic features, acting via different mechanisms. Both are potentially useful to specifically down‐regulate unwanted immune responses and induce immune tolerance. These modulated DC appear suitable as adjuvant in antigen‐specific clinical vaccination intervention strategies.


Nature Medicine | 2006

Selective cytotoxic T-lymphocyte targeting of tumor immune escape variants

Thorbald van Hall; Elisabeth Z. Wolpert; Peter A. van Veelen; Sandra Laban; Michael van der Veer; Marjet Roseboom; Sandra A. Bres; Per Grufman; Arnoud H. de Ru; Hugo D. Meiring; Ad P. J. M. de Jong; Kees L. M. C. Franken; Antoinette Teixeira; Rob Valentijn; Jan W. Drijfhout; Frits Koning; Marcel Camps; Ferry Ossendorp; Klas Kärre; Hans-Gustaf Ljunggren; Cornelis J. M. Melief; Rienk Offringa

Defects in major histocompatibility complex (MHC) class I–restricted antigen presentation are frequently observed in human cancers and result in escape of tumors from cytotoxic T lymphocyte (CTL) immune surveillance in mice. Here, we show the existence of a unique category of CTLs that can prevent this escape. The CTLs target an alternative repertoire of peptide epitopes that emerge in MHC class I at the surface of cells with impaired function of transporter associated with antigen processing (TAP), tapasin or the proteasome. These peptides, although derived from self antigens such as the commonly expressed Lass5 protein (also known as Trh4), are not presented by normal cells. This explains why they act as immunogenic neoantigens. The newly discovered epitopes can be exploited for immune intervention against processing-deficient tumors through adoptive T-cell transfer or peptide vaccination.


Journal of Immunology | 2010

Critical Role for TNF in the Induction of Human Antigen-Specific Regulatory T Cells by Tolerogenic Dendritic Cells

Fleur S. Kleijwegt; Sandra Laban; Gaby Duinkerken; Antoinette M. Joosten; Arnaud Zaldumbide; Tatjana Nikolic; Bart O. Roep

TNF is a pleiotropic cytokine with differential effects on immune cells and diseases. Anti-TNF therapy was shown to be effective in rheumatoid arthritis but proved inefficient or even detrimental in other autoimmune diseases. We studied the role of TNF in the induction of Ag-specific regulatory T cells (Tregs) by tolerogenic vitamin D3-modulated human dendritic cells (VD3-DCs), which previously were shown to release high amounts of soluble TNF (sTNF) upon maturation with LPS. First, production of TNF by modulated VD3-DCs was analyzed upon maturation with LPS or CD40L with respect to both secreted (cleaved) TNF (sTNF) and expression of the membrane-bound (uncleaved) form of TNF (mTNF). Next, TNF antagonists were tested for their effect on induction of Ag-specific Tregs by modulated DCs and the subsequent functionality of these Tregs. VD3-DCs expressed greater amounts of mTNF than did control DCs (nontreated DCs), independent of the maturation protocol. Inhibition of TNF with anti-TNF Ab (blocking both sTNF and mTNF) during the priming of Tregs with VD3-DCs prevented generation of Tregs and their suppression of proliferation of CD4+ T cells. In contrast, sTNF receptor II (sTNFRII), mainly blocking sTNF, did not change the suppressive capacity of Tregs. Blocking of TNFRII by anti-CD120b Ab during Treg induction similarly abrogated their subsequent suppressive function. These data point to a specific role for mTNF on VD3-DCs in the induction of Ag-specific Tregs. Interaction between mTNF and TNFRII instructs the induction of suppressive Tregs by VD3-DCs. Anti-TNF therapy may therefore act adversely in different patients or disease pathways.


Transplantation | 2003

Prolongation of skin graft survival by modulation of the alloimmune response with alternatively activated dendritic cells.

Dave L. Roelen; Danita H. Schuurhuis; Dani lle E. M. van den Boogaardt; Karin Koekkoek; Paula P.M.C. van Miert; Jolien J. van chip; Sandra Laban; Delphine Rea; Cees J. M. Melief; Rienk Offringa; Ferry Ossendorp; Frans H.J. Claas

Background. Activation of immature dendritic cells (DC) in the presence of the glucocorticoid hormone dexamethasone (DEX) results in alternatively matured DC that present antigen in the absence of a proper co-stimulatory context. This maturation process is irreversible, making these cells an attractive potential tool for the induction of antigen-specific T-cell tolerance in vivo. The authors explored the possibility of using these DC for the induction of transplantation tolerance in a fully allogeneic setting in mice. Methods. Immature dendritic cells (D1, an immature splenic DC line derived from B6 mice) were pretreated with DEX for 24 hr, after which lipopolysaccharide or nothing was added to the culture for another 48 hr. These cells were analyzed for their in vitro and in vivo stimulating or tolerizing capacities. Results. In line with their phenotype, including decreased interleukin (IL)-12 production, in vitro co-culture of alternatively matured D1 (B6 origin; H-2b) with completely allogeneic T cells of BALB/c origin led to a significant decrease in the alloreactive T-cell response. A single injection of 1×106 alternatively matured H-2b DC into BALB/c mice induced a different alloimmune response compared with mature DC. The responding T cells showed a lower proliferation rate and a lower interferon-&ggr; production, whereas a significantly higher proportion of the cells produced IL-10 as measured ex vivo by enzyme-linked immunospot assay. Furthermore, injection with alternatively matured DC, followed by transplantation of fully mismatched skin grafts (C57BL/6), led to a significantly prolonged survival compared with that of mature DC-pretreated mice or untreated mice. The immunomodulatory effect was antigen specific, as third-party reactive alloresponses were not affected. Conclusions. The authors’ data constitute the first direct demonstration that DC alternatively matured in the presence of glucocorticoid hormones can be exploited for the specific suppression of the alloreactive Th1 response, resulting in a delayed skin graft rejection in a complete major histocompatibility complex-incompatible strain combination.


Journal of Immunology | 2009

TLR Triggering on Tolerogenic Dendritic Cells Results in TLR2 Up-Regulation and a Reduced Proinflammatory Immune Program

Sonia Chamorro; Juan J. Garcia-Vallejo; Wendy W. J. Unger; Rosette J. Fernandes; Sven C. M. Bruijns; Sandra Laban; Bart O. Roep; Bert A. 't Hart; Yvette van Kooyk

Tolerogenic dendritic cells (TDC) offer a promising therapeutic potential to ameliorate autoimmune diseases. Reported to inhibit adaptive immune responses, little is known about their innate immunity receptor repertoire. In this study, we compared three types of human TDC (IL-10-DC, dexamethasone (DX)-DC, and 1,25(OH)2D3-DC) by their TLR expression and response to a set of TLR ligands. TDC are endowed with the same TLR set as standard monocyte-derived dendritic cells but respond differentially to the TLR stimuli Pam3CSK4, polyinosinic-polycytidylic acid, LPS, and flagellin. TDC expressed low or no IL-12-related cytokines and remarkably elevated IL-10 levels. Interestingly, only TDC up-regulated the expression of TLR2 upon stimulation. This boosted the tolerogenic potential of these cells, because IL-10 production was up-regulated in TLR2-stimulated, LPS-primed DX-DC, whereas IL-12 and TNF-α secretion remained low. When comparing the TDC subsets, DX-DC and 1,25(OH)2D3-DC up-regulated TLR2 irrespective of the TLR triggered, whereas in IL-10-DC this effect was only mediated by LPS. Likewise, DX-DC and 1,25(OH)2D3-DC exhibited impaired ability to mature, reduced allostimulatory properties, and hampered capacity to induce Th1 differentiation. Therefore, both DX-DC and 1,25(OH)2D3-DC display the strongest tolerogenic and anti-inflammatory features and might be most suitable tools for the treatment of autoimmune diseases.


PLOS ONE | 2012

Islet-Specific CTL Cloned from a Type 1 Diabetes Patient Cause Beta-Cell Destruction after Engraftment into HLA-A2 Transgenic NOD/SCID/IL2RG Null Mice

Wendy W. J. Unger; Todd Pearson; Joana R. F. Abreu; Sandra Laban; Arno R. van der Slik; Sacha Mulder-van der Kracht; Michel G.D. Kester; Dave V. Serreze; Leonard D. Shultz; Marieke Griffioen; Jan W. Drijfhout; Dale L. Greiner; Bart O. Roep

Despite increasing evidence that autoreactive CD8 T-cells are involved in both the initiation of type 1 diabetes (T1D) and the destruction of beta-cells, direct evidence for their destructive role in-vivo is lacking. To address a destructive role for autoreactive CD8 T-cells in human disease, we assessed the pathogenicity of a CD8 T-cell clone derived from a T1D donor and specific for an HLA-A2-restricted epitope of islet-specific glucose-6-phosphatase catalytic-subunit related protein (IGRP). HLA-A2/IGRP tetramer staining revealed a higher frequency of IGRP-specific CD8 T-cells in the peripheral blood of recent onset human individuals than of healthy donors. IGRP265–273-specific CD8 T-cells that were cloned from the peripheral blood of a recent onset T1D individual were shown to secrete IFNγ and Granzyme B after antigen-specific activation and lyse HLA-A2-expressing murine islets in-vitro. Lytic capacity was also demonstrated in-vivo by specific killing of peptide-pulsed target cells. Using the HLA-A2 NOD-scid IL2rγnull mouse model, HLA-A2-restricted IGRP-specific CD8 T-cells induced a destructive insulitis. Together, this is the first evidence that human HLA-restricted autoreactive CD8 T-cells target HLA-expressing beta-cells in-vivo, demonstrating the translational value of humanized mice to study mechanisms of disease and therapeutic intervention strategies.


Nature Medicine | 2017

Autoimmunity against a defective ribosomal insulin gene product in type 1 diabetes

Maria Jl Kracht; Menno van Lummel; Tatjana Nikolic; Antoinette M. Joosten; Sandra Laban; Arno R. van der Slik; Peter A. van Veelen; Françoise Carlotti; Eelco J.P. de Koning; Rob C. Hoeben; Arnaud Zaldumbide; Bart O. Roep

Identification of epitopes that are recognized by diabetogenic T cells and cause selective beta cell destruction in type 1 diabetes (T1D) has focused on peptides originating from native beta cell proteins. Translational errors represent a major potential source of antigenic peptides to which central immune tolerance is lacking. Here, we describe an alternative open reading frame within human insulin mRNA encoding a highly immunogenic polypeptide that is targeted by T cells in T1D patients. We show that cytotoxic T cells directed against the N-terminal peptide of this nonconventional product are present in the circulation of individuals diagnosed with T1D, and we provide direct evidence that such CD8+ T cells are capable of killing human beta cells and thereby may be diabetogenic. This study reveals a new source of nonconventional polypeptides that act as self-epitopes in clinical autoimmune disease.


Journal of Autoimmunity | 2011

Discovery of low-affinity preproinsulin epitopes and detection of autoreactive CD8 T-cells using combinatorial MHC multimers.

Wendy W. J. Unger; Jurjen Velthuis; Joana R. F. Abreu; Sandra Laban; Edwin Quinten; Michel G.D. Kester; Sine Reker-Hadrup; Arnold H. Bakker; Gaby Duinkerken; Arend Mulder; Kees L. M. C. Franken; Robert Hilbrands; Bart Keymeulen; Mark Peakman; Ferry Ossendorp; Jan W. Drijfhout; Ton N. M. Schumacher; Bart O. Roep

Autoreactive cytotoxic CD8 T-cells (CTLs) play a key pathogenic role in the destruction of insulin-producing beta-cells resulting in type 1 diabetes. However, knowledge regarding their targets is limited, restricting the ability to monitor the course of the disease and immune interventions. In a multi-step discovery process to identify novel CTL epitopes in human preproinsulin (PPI), PPI was digested with purified human proteasomes, and resulting COOH-fragments aligned with algorithm-predicted HLA-binding peptides to yield nine potential HLA-A1, -A2, -A3 or -B7-restricted candidates. An UV-exchange method allowed the generation of a repertoire of multimers including low-affinity HLA-binding peptides. These were labeled with quantum dot-fluorochromes and encoded in a combinatorial fashion, allowing parallel and sensitive detection of specific, low-avidity T-cells. Significantly increased frequencies of T-cells against four novel PPI epitopes (PPI(4-13)/B7, PPI(29-38)/A2, PPI(76-84)/A3 and PPI(79-88)/A3) were detected in stored blood of patients with recent onset diabetes but not in controls. Changes in frequencies of circulating CD8 T-cells against these novel epitopes were detected in blood of islet graft recipients at different time points after transplantation, which correlated with clinical outcome. In conclusion, our novel strategy involving a sensitive multiplex detection technology and requiring minimal volumes of stored blood represents a major improvement in the direct ex-vivo characterization and enumeration of immune cells in the pathogenesis of type 1 diabetes.


Journal of Immunology | 2007

The Varicellovirus-Encoded TAP Inhibitor UL49.5 Regulates the Presentation of CTL Epitopes by Qa-1b1

Thorbald van Hall; Sandra Laban; Danijela Koppers-Lalic; Joachim Koch; Calin Precup; Patrick S. Asmawidjaja; Rienk Offringa; Emmanuel J. H. J. Wiertz

Impairment of MHC class I Ag processing is a commonly observed mechanism that allows viruses and tumors to escape immune destruction by CTL. The peptide transporter TAP that is responsible for the delivery of MHC class I-binding peptides into the endoplasmic reticulum is a pivotal target of viral-immune evasion molecules, and expression of this transporter is frequently lost in advanced cancers. We recently described a novel population of CTL that intriguingly exhibits reactivity against such tumor-immune escape variants and that recognizes self-peptides emerging at the cell surface due to defects in the processing machinery. Investigations of this new type of CTL epitopes are hampered by the lack of an efficient inhibitor for peptide transport in mouse cells. In this article, we demonstrate that the varicellovirus protein UL49.5, in contrast to ICP47 and US6, strongly impairs the activity of the mouse transporter and mediates degradation of mouse TAP1 and TAP2. Inhibition of TAP was witnessed by a strong reduction of surface MHC class I display and a decrease in recognition of conventional tumor-specific CTL. Analysis of CTL reactivity through the nonclassical molecule Qa-1b revealed that the presentation of the predominant leader peptide was inhibited. Interestingly, expression of UL49.5 in processing competent tumor cells induced the presentation of the new category of peptides. Our data show that the varicellovirus UL49.5 protein is a universal TAP inhibitor that can be exploited for preclinical studies on CTL-based immune intervention.


Journal of Immunology | 2011

Transfer of Regulatory Properties from Tolerogenic to Proinflammatory Dendritic Cells via Induced Autoreactive Regulatory T Cells

Fleur S. Kleijwegt; Sandra Laban; Gaby Duinkerken; Antoinette M. Joosten; Bobby P. C. Koeleman; Tatjana Nikolic; Bart O. Roep

Infectious tolerance is a term generally assigned to the process through which regulatory T cells (Tregs) transfer immunoregulatory properties to other T cells. In this study, we demonstrated that a similar process applies to human dendritic cells (DCs), albeit through a different mechanism. We induced and cloned proinsulin-specific Tregs using tolerogenic DCs and investigated mechanisms by which induced Ag-specific regulatory T cells (iaTregs) endorse the suppressive effects. iaTregs expressed FOXP3, programmed death-1, and membrane-bound TGF-β and upregulated IL-10 and CTLA-4 after stimulation with the cognate Ag. The iaTregs suppressed effector T cells only when both encountered the cognate Ags on the same APCs (linked suppression). This occurred independently of IL-10, TGF-β, programmed death-1, or CTLA-4. Instead, iaTregs used a granzyme B-mediated mechanism to kill B cells and monocytes, whereas proinflammatory DCs that resisted being killed were induced to upregulate the inhibitory receptors B7 (family) homolog 3 and ICOS ligand. These re-educated mature monocyte-derived dendritic cells (mDCs) suppressed effector T cells and induced IL-10–producing cells from the naive T cell pool. Our data indicated that human tolerogenic DCs confer infectious tolerance by inducing Ag-specific Tregs, which, in turn, re-educate proinflammatory mature DCs into DCs with regulatory properties.

Collaboration


Dive into the Sandra Laban's collaboration.

Top Co-Authors

Avatar

Bart O. Roep

Beckman Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tatjana Nikolic

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rienk Offringa

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan W. Drijfhout

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arnaud Zaldumbide

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ferry Ossendorp

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter A. van Veelen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arno R. van der Slik

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge