Network


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

Hotspot


Dive into the research topics where Tatjana Nikolic is active.

Publication


Featured researches published by Tatjana Nikolic.


Journal of Proteome Research | 2012

Differential Protein Pathways in 1,25-Dihydroxyvitamin D-3 and Dexamethasone Modulated Tolerogenic Human Dendritic Cells

Gabriela B Ferreira; Fleur S. Kleijwegt; Etienne Waelkens; Kasper Lage; Tatjana Nikolic; Daniel Aaen Hansen; Christopher T. Workman; Bart O. Roep; Lut Overbergh; Chantal Mathieu

Tolerogenic dendritic cells (DC) that are maturation-resistant and locked in a semimature state are promising tools in clinical applications for tolerance induction. Different immunomodulatory agents have been shown to induce a tolerogenic DC phenotype, such as the biologically active form of vitamin D (1,25(OH)(2)D(3)), glucocorticoids, and a synergistic combination of both. In this study, we aimed to characterize the protein profile, function and phenotype of DCs obtained in vitro in the presence of 1,25(OH)(2)D(3), dexamethasone (DEX), and a combination of both compounds (combi). Human CD14(+) monocytes were differentiated toward mature DCs, in the presence or absence of 1,25(OH)(2)D(3) and/or DEX. Cells were prefractionated into cytoplasmic and microsomal fractions and protein samples were separated in two different pH ranges (pH 3-7NL and 6-9), analyzed by 2D-DIGE and differentially expressed spots (p < 0.05) were identified after MALDI-TOF/TOF analysis. In parallel, morphological and phenotypical analyses were performed, revealing that 1,25(OH)(2)D(3)- and combi-mDCs are closer related to each other than DEX-mDCs. This was translated in their protein profile, indicating that 1,25(OH)(2)D(3) is more potent than DEX in inducing a tolerogenic profile on human DCs. Moreover, we demonstrate that combining 1,25(OH)(2)D(3) with DEX induces a unique protein expression pattern with major imprinting of the 1,25(OH)(2)D(3) effect. Finally, protein interaction networks and pathway analysis suggest that 1,25(OH)(2)D(3), rather than DEX treatment, has a severe impact on metabolic pathways involving lipids, glucose, and oxidative phosphorylation, which may affect the production of or the response to ROS generation. These findings provide new insights on the molecular basis of DC tolerogenicity induced by 1,25(OH)(2)D(3) and/or DEX, which may lead to the discovery of new pathways involved in DC immunomodulation.


Frontiers in Immunology | 2013

Regulatory multitasking of tolerogenic dendritic cells - lessons taken from vitamin d3-treated tolerogenic dendritic cells.

Tatjana Nikolic; Bart O. Roep

Tolerogenic dendritic cells (DCs) work through silencing of differentiated antigen-specific T cells, activation and expansion of naturally occurring T regulatory cells (Tregs), transfer of regulatory properties to T cells, and the differentiation of naïve T cells into Tregs. Due to an operational definition based on T cell activation assays, the identity of tolerogenic DCs has been a matter of debate and it need not represent a specialized DC subset. Human tolerogenic DCs generated in vitro using inhibitory cytokines, growth factors, natural immunomodulators, or genetic manipulation have been effective and several of these tolerogenic DCs are currently being tested for clinical use. Ex vivo generated tolerogenic DCs reduce activation of naïve T cells using various means, promote a variety of regulatory T cells and most importantly, frequently show stable inhibitory phenotypes upon repetitive maturation with inflammatory factors. Yet, tolerogenic DCs differ with respect to the phenotype or the number of regulatory mechanisms they employ to modulate the immune system. In our experience, tolerogenic DCs generated using the biologically active form of vitamin D (VD3-DCs), alone, or combined with dexamethasone are proficient in their immunoregulatory functions. These tolerogenic DCs show a stable maturation-resistant semi-mature phenotype with low expression of activating co-stimulatory molecules, no production of the IL-12 family of cytokines and high expression of inhibitory molecules and IL-10. VD3-DCs induce increased apoptosis of effector T cells and induce antigen-specific regulatory T cells, which work through linked suppression ensuring infectious tolerance. Lessons learned on VD3-DCs help understanding the contribution of different pattern-recognition receptors (PRRs) and secondary signals to the tolerogenic function and how a cross-talk between DCs and T cells translates into immune regulation.


Nature Immunology | 2015

T cell receptor reversed polarity recognition of a self-antigen major histocompatibility complex.

Dennis X. Beringer; Fleur S. Kleijwegt; Arno R. van der Slik; Khai Lee Loh; Jan Petersen; Nadine L. Dudek; Gaby Duinkerken; Sandra Laban; Antoinette M. Joosten; Julian P. Vivian; Zhenjun Chen; Anthony W. Uldrich; Dale I. Godfrey; James McCluskey; David A. Price; Kristen J. Radford; Anthony W. Purcell; Tatjana Nikolic; Hugh H. Reid; Tony Tiganis; Bart O. Roep; Jamie Rossjohn

Central to adaptive immunity is the interaction between the αβ T cell receptor (TCR) and peptide presented by the major histocompatibility complex (MHC) molecule. Presumably reflecting TCR-MHC bias and T cell signaling constraints, the TCR universally adopts a canonical polarity atop the MHC. We report the structures of two TCRs, derived from human induced T regulatory (iTreg) cells, complexed to an MHC class II molecule presenting a proinsulin-derived peptide. The ternary complexes revealed a 180° polarity reversal compared to all other TCR-peptide-MHC complex structures. Namely, the iTreg TCR α-chain and β-chain are overlaid with the α-chain and β-chain of MHC class II, respectively. Nevertheless, this TCR interaction elicited a peptide-reactive, MHC-restricted T cell signal. Thus TCRs are not hardwired to interact with MHC molecules in a stereotypic manner to elicit a T cell signal, a finding that fundamentally challenges our understanding of TCR recognition.


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 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.


European Journal of Immunology | 2013

Tolerogenic dendritic cells impede priming of naïve CD8+ T cells and deplete memory CD8+ T cells

Fleur S. Kleijwegt; Diahann T. S. L. Jansen; Josefine Teeler; Antoinette M. Joosten; Sandra Laban; Tatjana Nikolic; Bart O. Roep

Type 1 diabetes is a T‐cell‐mediated autoimmune disease in which autoreactive CD8+ T cells destroy the insulin‐producing pancreatic beta cells. Vitamin D3 and dexamethasone‐modulated dendritic cells (Combi‐DCs) loaded with islet antigens inducing islet‐specific regulatory CD4+ T cells may offer a tissue‐specific intervention therapy. The effect of Combi‐DCs on CD8+ T cells, however, remains unknown. To investigate the interaction of CD8+ T cells with Combi‐DCs presenting epitopes on HLA class I, naive, and memory CD8+ T cells were co‐cultured with DCs and proliferation and function of peptide‐specific T cells were analyzed. Antigen‐loaded Combi‐DCs were unable to prime naïve CD8+ T cells to proliferate, although a proportion of T cells converted to a memory phenotype. Moreover, expansion of CD8+ T cells that had been primed by mature monocyte‐derived DCs (moDCs) was curtailed by Combi‐DCs in co‐cultures. Combi‐DCs expanded memory T cells once, but CD8+ T‐cell numbers collapsed by subsequent re‐stimulation with Combi‐DCs. Our data point that (re)activation of CD8+ T cells by antigen‐pulsed Combi‐DCs does not promote, but rather deteriorates, CD8+ T‐cell immunity. Yet, Combi‐DCs pulsed with CD8+ T‐cell epitopes also act as targets of cytotoxicity, which is undesirable for survival of Combi‐DCs infused into patients in therapeutic immune intervention strategies.


Clinical and Experimental Immunology | 2015

Proinsulin multi-peptide immunotherapy induces antigen-specific regulatory T cells and limits autoimmunity in a humanized model

Vivienne Gibson; Tatjana Nikolic; Verity Pearce; J. Demengeot; Bart O. Roep; Mark Peakman

Peptide immunotherapy (PIT) is a targeted therapeutic approach, involving administration of disease‐associated peptides, with the aim of restoring antigen‐specific immunological tolerance without generalized immunosuppression. In type 1 diabetes, proinsulin is a primary antigen targeted by the autoimmune response, and is therefore a strong candidate for exploitation via PIT in this setting. To elucidate the optimal conditions for proinsulin‐based PIT and explore mechanisms of action, we developed a preclinical model of proinsulin autoimmunity in a humanized HLA‐DRB1*0401 transgenic HLA‐DR4 Tg mouse. Once proinsulin‐specific tolerance is broken, HLA‐DR4 Tg mice develop autoinflammatory responses, including proinsulin‐specific T cell proliferation, interferon (IFN)‐γ and autoantibody production. These are preventable and quenchable by pre‐ and post‐induction treatment, respectively, using intradermal proinsulin‐PIT injections. Intradermal proinsulin‐PIT enhances proliferation of regulatory [forkhead box protein 3 (FoxP3+)CD25high] CD4 T cells, including those capable of proinsulin‐specific regulation, suggesting this as its main mode of action. In contrast, peptide delivered intradermally on the surface of vitamin D3‐modulated (tolerogenic) dendritic cells, controls autoimmunity in association with proinsulin‐specific IL‐10 production, but no change in regulatory CD4 T cells. These studies define a humanized, translational model for in vivo optimization of PIT to control autoimmunity in type 1 diabetes and indicate that dominant mechanisms of action differ according to mode of peptide delivery.


PeerJ | 2016

Minimum information about tolerogenic antigen-presenting cells (MITAP) : a first step towards reproducibility and standardisation of cellular therapies

Phillip Lord; Rachel Spiering; Juan Carlos Aguillón; Amy E. Anderson; Silke Appel; Daniel Benitez-Ribas; Anja ten Brinke; Femke Broere; Nathalie Cools; Maria Cristina Cuturi; Julie Diboll; Edward K. Geissler; Nick Giannoukakis; Silvia Gregori; S. Marieke van Ham; Staci Lattimer; Lindsay Marshall; Rachel A. Harry; James A. Hutchinson; John D. Isaacs; Irma Joosten; Cees van Kooten; Ascension Lopez Diaz de Cerio; Tatjana Nikolic; Haluk Barbaros Oral; Ljiljana Sofronic-Milosavljevic; Thomas Ritter; Paloma Riquelme; Angus W. Thomson; Massimo Trucco

Cellular therapies with tolerogenic antigen-presenting cells (tolAPC) show great promise for the treatment of autoimmune diseases and for the prevention of destructive immune responses after transplantation. The methodologies for generating tolAPC vary greatly between different laboratories, making it difficult to compare data from different studies; thus constituting a major hurdle for the development of standardised tolAPC therapeutic products. Here we describe an initiative by members of the tolAPC field to generate a minimum information model for tolAPC (MITAP), providing a reporting framework that will make differences and similarities between tolAPC products transparent. In this way, MITAP constitutes a first but important step towards the production of standardised and reproducible tolAPC for clinical application.


Diabetes | 2016

Discovery of a selective islet peptidome presented by the highest-risk HLA-DQ8trans molecule

Menno van Lummel; Peter A. van Veelen; Arnoud H. de Ru; Jos Pool; Tatjana Nikolic; Sandra Laban; Antoinette M. Joosten; Jan W. Drijfhout; Iria Gómez-Touriño; Sefina Arif; Henk J. Aanstoot; Mark Peakman; Bart O. Roep

HLA-DQ2/8 heterozygous individuals are at far greater risk for type 1 diabetes (T1D) development by expressing HLA-DQ8trans on antigen-presenting cells compared with HLA-DQ2 or -DQ8 homozygous individuals. Dendritic cells (DC) initiate and shape adaptive immune responses by presenting HLA-epitope complexes to naïve T cells. To dissect the role of HLA-DQ8trans in presenting natural islet epitopes, we analyzed the islet peptidome of HLA-DQ2, -DQ8, and -DQ2/8 by pulsing DC with preproinsulin (PPI), IA-2, and GAD65. Quality and quantity of islet epitopes presented by HLA-DQ2/8 differed from -DQ2 or -DQ8. We identified two PPI epitopes solely processed and presented by HLA-DQ2/8 DC: an HLA-DQ8trans–binding signal-sequence epitope previously identified as CD8 T-cell epitope and a second epitope that we previously identified as CD4 T-cell epitope with increased binding to HLA-DQ8trans upon posttranslational modification. IA-2 epitopes retrieved from HLA-DQ2/8 and -DQ8 DC bound to HLA-DQ8cis/trans. No GAD65 epitopes were eluted from HLA-DQ. T-cell responses were detected against the novel islet epitopes in blood from patients with T1D but scantly detected in healthy donor subjects. We report the first PPI and IA-2 natural epitopes presented by highest-risk HLA-DQ8trans. The selective processing and presentation of HLA-DQ8trans–binding islet epitopes provides insight in the mechanism of excessive genetic risk imposed by HLA-DQ2/8 heterozygosity and may assist immune monitoring of disease progression and therapeutic intervention as well as provide therapeutic targets for immunotherapy in subjects at risk for T1D.


Molecular Therapy | 2013

Genetically engineered human islets protected from CD8-mediated autoimmune destruction in vivo.

Arnaud Zaldumbide; Gonnie M. Alkemade; Françoise Carlotti; Tatjana Nikolic; Joana R. F. Abreu; Marten A. Engelse; Anja Skowera; Eelco J.P. de Koning; Mark Peakman; Bart O. Roep; Rob C. Hoeben; Emmanuel J. H. J. Wiertz

Islet transplantation is a promising therapy for type 1 diabetes, but graft function and survival are compromised by recurrent islet autoimmunity. Immunoprotection of islets will be required to improve clinical outcome. We engineered human β cells to express herpesvirus-encoded immune-evasion proteins, immunevasins. The capacity of immunevasins to protect β cells from autoreactive T-cell killing was evaluated in vitro and in vivo in humanized mice. Lentiviral vectors were used for efficient genetic modification of primary human β cells without impairing their function. Using a novel β-cell-specific reporter gene assay, we show that autoreactive cytotoxic CD8(+) T-cell clones isolated from patients with recent onset diabetes selectively destroyed human β cells, and that coexpression of the human cytomegalovirus-encoded US2 protein and serine proteinase inhibitor 9 offers highly efficient protection in vitro. Moreover, coimplantation of these genetically modified pseudoislets with β-cell-specific cytotoxic T cells into immunodeficient mice achieves preserved human insulin production and C-peptide secretion. Collectively, our data provide proof of concept that human β cells can be efficiently genetically modified to provide protection from killing mediated by autoreactive T cells and retain their function in vitro and in vivo.

Collaboration


Dive into the Tatjana Nikolic's collaboration.

Top Co-Authors

Avatar

Bart O. Roep

Beckman Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandra Laban

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Menno van Lummel

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

Arnaud Zaldumbide

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
Top Co-Authors

Avatar

Arnoud H. de Ru

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge