Network


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

Hotspot


Dive into the research topics where Ingrid A. M. Derks is active.

Publication


Featured researches published by Ingrid A. M. Derks.


Immunity | 2010

Apoptosis Threshold Set by Noxa and Mcl-1 after T Cell Activation Regulates Competitive Selection of High-Affinity Clones

Felix M. Wensveen; Klaas P. J. M. van Gisbergen; Ingrid A. M. Derks; Carmen Gerlach; Ton N. M. Schumacher; René A. W. van Lier; Eric Eldering

The adaptive immune system generates protective T cell responses via a poorly understood selection mechanism that favors expansion of clones with optimal affinity for antigen. Here we showed that upon T cell activation, the proapoptotic molecule Noxa (encoded by Pmaip1) and its antagonist Mcl-1 were induced. During an acute immune response against influenza or ovalbumin, Pmaip1(-/-) effector T cells displayed decreased antigen affinity and functionality. Molecular analysis of influenza-specific T cells revealed persistence of many subdominant clones in the Pmaip1(-/-) effector pool. When competing for low-affinity antigen, Pmaip1(-/-) TCR transgenic T cells had a survival advantage in vitro, resulting in increased numbers of effector cells in vivo. Mcl-1 protein stability was controlled by T cell receptor (TCR) affinity-dependent interleukin-2 signaling. These results establish a role for apoptosis early during T cell expansion, based on antigen-driven competition and survival of the fittest T cells.


Journal of Immunology | 2008

Differential regulation of human IL-7 receptor alpha expression by IL-7 and TCR signaling.

Nuno L. Alves; Ester M. M. van Leeuwen; Ingrid A. M. Derks; René A. W. van Lier

IL-7Rα is essential for the development and homeostatic maintenance of mature T cells. Studies in humans and mice have shown that IL-7Rα expression is reduced by its cognate cytokine, IL-7, and Ag, suggesting that active regulation of IL-7 responsiveness is necessary to balance T cell numbers. We show that IL-7- or TCR/CD28-mediated signaling induced a rapid down-regulation of IL-7Rα expression on naive T cells on the mRNA and protein level, with a mild (10-fold) or strong (50-fold) gene suppression, respectively. In both situations, the down-regulation of IL-7Rα was blocked by cyclohexamide and actinomycin D, indicating the involvement of an active mechanism dependent on new transcription and protein synthesis. Upon IL-7 withdrawal, IL-7Rα mRNA and surface protein reappeared in a transcription-dependent manner within 7 h. Yet, IL-7Rα was hardly re-expressed during the same period after TCR/CD28-activation. Likewise, T cells that were activated through CMV in vivo did not re-express IL-7Rα after in vitro culture. Functionally, IL-7-induced down-regulation of IL-7Rα did not hinder the responsiveness of naive T cells to IL-7. Conversely, down-regulation of IL-7Rα on TCR/CD28-activated cells limited IL-7 responsiveness. Strikingly, ectopic expression of IL-7Rα cells on TCR/CD28-activated cells conferred a selective advantage in the response to IL-7. In conclusion, our data show that IL-7- and TCR/CD28-mediated signaling differentially regulate IL-7Rα expression on human T cells with a transient and chronic effect, respectively. The stringent and active regulation of IL-7Rα may constitute a homeostatic mechanism to curtail unwarranted T cell expansion.


Blood | 2013

IL-21 and CD40L signals from autologous T cells can induce antigen-independent proliferation of CLL cells

Maria Fernanda Pascutti; Margot Jak; Jacqueline M. Tromp; Ingrid A. M. Derks; Ester B. M. Remmerswaal; Rachel Thijssen; Martijn H. A. van Attekum; Gregor van Bochove; Dieuwertje M. Luijks; Steven T. Pals; René A. W. van Lier; Arnon P. Kater; Marinus H. J. van Oers; Eric Eldering

Chronic lymphocytic leukemia (CLL) cells multiply in secondary lymphoid tissue, but the mechanisms leading to their proliferation are still uncertain. In addition to B-cell receptor (BCR)-triggered signals, other microenvironmental factors might well be involved. In proliferation centers, leukemic B cells are in close contact with CD4(+)CD40L(+) T cells. Therefore, we here dissected the signals provided by autologous activated T cells (Tact) to CLL cells. Although the gene expression profile induced by Tact was highly similar to that induced by sole CD40 signaling, an obvious difference was that Tact induced proliferation of CLL cells. We determined that stimulation with only CD40L+IL-21 was sufficient to induce robust proliferation in CLL cells. We then defined an interleukin (IL)-21-induced gene signature in CLL, containing components of Janus kinase/signal transducer and activator of transcription and apoptosis pathways, and this signature could be detected in lymph node (LN) samples from patients. Finally, we could detect IL-21 RNA and protein in LN, and IL-21 production ex vivo by LN CD4(+)CXCR5(+) follicular helper T cells. These results indicate that in addition to BCR signaling, activated T cells might contribute to CLL cell proliferation via CD40 and IL-21. Targeting these signaling pathways might offer new venues for treatment of CLL.


Cell Death & Differentiation | 2007

Crosstalk among Bcl-2 family members in B-CLL: seliciclib acts via the Mcl-1/Noxa axis and gradual exhaustion of Bcl-2 protection.

Delfine Y.H. Hallaert; René Spijker; Margot Jak; Ingrid A. M. Derks; Nuno L. Alves; F M Wensveen; J de Boer; D. de Jong; Simon Green; M H J van Oers; E Eldering

Seliciclib (R-roscovitine) is a cyclin-dependent kinase inhibitor in clinical development. It triggers apoptosis by inhibiting de novo transcription of the short-lived Mcl-1 protein, but it is unknown how this leads to Bax/Bak activation that is required for most forms of cell death. Here, we studied the effects of seliciclib in B-cell chronic lymphocytic leukemia (B-CLL), a malignancy with aberrant expression of apoptosis regulators. Although seliciclib-induced Mcl-1 degradation within 4 h, Bax/Bak activation occurred between 16 and 20 h. During this period, no transcriptional changes in apoptosis-related genes occurred. In untreated cells, prosurvival Mcl-1 was engaged by the proapoptotic proteins Noxa and Bim. Upon drug treatment, Bim was quickly released. The contribution of Noxa and Bim as a specific mediator of seliciclib-induced apoptosis was demonstrated via RNAi. Significantly, 16 h after seliciclib treatment, there was accumulation of Bcl-2, Bim and Bax in the ‘mitochondria-rich’ insoluble fraction of the cell. This suggests that after Mcl-1 degradation, the remaining apoptosis neutralizing capacity of Bcl-2 is gradually overwhelmed, until Bax forms large multimeric pores in the mitochondria. These data demonstrate in primary leukemic cells hierarchical binding and crosstalk among Bcl-2 members, and suggest that their functional interdependence can be exploited therapeutically.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Notch controls the magnitude of T helper cell responses by promoting cellular longevity

Christina Helbig; Rebecca Gentek; Ronald Backer; Yevan de Souza; Ingrid A. M. Derks; Eric Eldering; Koen Wagner; Dragana Jankovic; Thomas Gridley; Perry D. Moerland; Richard A. Flavell; Derk Amsen

Generation of effective immune responses requires expansion of rare antigen-specific CD4+ T cells. The magnitude of the responding population is ultimately determined by proliferation and survival. Both processes are tightly controlled to limit responses to innocuous antigens. Sustained expansion occurs only when innate immune sensors are activated by microbial stimuli or by adjuvants, which has important implications for vaccination. The molecular identity of the signals controlling sustained T-cell responses is not fully clear. Here, we describe a prominent role for the Notch pathway in this process. Coactivation of Notch allows accumulation of far greater numbers of activated CD4+ T cells than stimulation via T-cell receptor and classic costimulation alone. Notch does not overtly affect cell cycle entry or progression of CD4+ T cells. Instead, Notch protects activated CD4+ T cells against apoptosis after an initial phase of clonal expansion. Notch induces a broad antiapoptotic gene expression program that protects against intrinsic, as well as extrinsic, apoptosis pathways. Both Notch1 and Notch2 receptors and the canonical effector RBPJ (recombination signal binding protein for immunoglobulin kappa J region) are involved in this process. Correspondingly, CD4+ T-cell responses to immunization with protein antigen are strongly reduced in mice lacking these components of the Notch pathway. Our findings, therefore, show that Notch controls the magnitude of CD4+ T-cell responses by promoting cellular longevity.


Apoptosis | 2011

Apoptosis induced by overall metabolic stress converges on the Bcl-2 family proteins Noxa and Mcl-1

Felix M. Wensveen; Nuno L. Alves; Ingrid A. M. Derks; Kris A. Reedquist; Eric Eldering

Apoptosis provoked by glucose shortage in dividing T cells is mediated via the BH3-only protein Noxa and inhibition of its binding partner Mcl-1. It is unknown how signals from cellular metabolism can affect the balance between Mcl-1 and Noxa and to what extent other Bcl-2 members are involved in this apoptosis cascade. Here, we defined the mechanism underlying apoptosis in relation to various types of metabolic stress. First, we established that the Noxa/Mcl-1 balance is regulated by glucose deprivation as well as by general metabolic stress, via changes in proteasome-mediated degradation of Mcl-1. Second, in contrast with cytokine-deprivation, no transcriptional modulation of Mcl-1, Puma, Bim or Noxa was observed during glucose deprivation. Third, no changes in PKB or GSK3 activity occurred and no clear role for AMPK was detected. Fourth, apoptosis triggered by nutrient deprivation was executed without signs of overt autophagy and independent of ROS production or p38 MAP kinase activity. Lastly, apoptosis under nutrient limitation could also be delayed by knock-down of Bim or overexpression of Bcl-2. In conclusion, Noxa functions in a specific apoptotic pathway that integrates overall nutrient stress, independent from attenuated PI3K/PKB signaling and without clear involvement of autophagy.


European Journal of Immunology | 2015

Blimp‐1 homolog Hobit identifies effector‐type lymphocytes in humans

Felipe A. Vieira Braga; Kirsten M. L. Hertoghs; Natasja A. M. Kragten; Gina M. Doody; Nicholas A. Barnes; Ester B. M. Remmerswaal; Cheng-Chih Hsiao; Perry D. Moerland; Diana Wouters; Ingrid A. M. Derks; Amber van Stijn; Marc Demkes; Jörg Hamann; Eric Eldering; Martijn A. Nolte; Reuben Tooze; Ineke J. M. ten Berge; Klaas P. J. M. van Gisbergen; René A. W. van Lier

Human cytomegalovirus (CMV) induces the formation of effector CD8+ T cells that are maintained for decades during the latent stage of infection. Effector CD8+ T cells appear quiescent, but maintain constitutive cytolytic capacity and can immediately produce inflammatory cytokines such as IFN‐γ after stimulation. It is unclear how effector CD8+ T cells can be constitutively maintained in a terminal stage of effector differentiation in the absence of overt viral replication. We have recently described the zinc finger protein Homolog of Blimp‐1 in T cells (Hobit) in murine NKT cells. Here, we show that human Hobit was uniformly expressed in effector‐type CD8+ T cells, but not in naive or in most memory CD8+ T cells. Human CMV‐specific but not influenza‐specific CD8+ T cells expressed high levels of Hobit. Consistent with the high homology between the DNA‐binding Zinc Finger domains of Hobit and Blimp‐1, Hobit displayed transcriptional activity at Blimp‐1 target sites. Expression of Hobit strongly correlated with T‐bet and IFN‐γ expression within the CD8+ T‐cell population. Furthermore, Hobit was both necessary and sufficient for the production of IFN‐γ. These data implicate Hobit as a novel transcriptional regulator in quiescent human effector‐type CD8+ T cells that regulates their immediate effector functions.


Leukemia | 2015

The impact of SF3B1 mutations in CLL on the DNA-damage response

G.D. te Raa; Ingrid A. M. Derks; Veronika Navrkalová; Anna Skowronska; Perry D. Moerland; J van Laar; Ceri Oldreive; H. Monsuur; Martin Trbušek; Jitka Malčíková; M. Lodén; Christian H Geisler; Jennifer Hüllein; Alexander Jethwa; T. Zenz; Šárka Pospíšilová; Tatjana Stankovic; M. H. J. Van Oers; Arnon P. Kater; E Eldering

Mutations or deletions in TP53 or ATM are well-known determinants of poor prognosis in chronic lymphocytic leukemia (CLL), but only account for approximately 40% of chemo-resistant patients. Genome-wide sequencing has uncovered novel mutations in the splicing factor sf3b1, that were in part associated with ATM aberrations, suggesting functional synergy. We first performed detailed genetic analyses in a CLL cohort (n=110) containing ATM, SF3B1 and TP53 gene defects. Next, we applied a newly developed multiplex assay for p53/ATM target gene induction and measured apoptotic responses to DNA damage. Interestingly, SF3B1 mutated samples without concurrent ATM and TP53 aberrations (sole SF3B1) displayed partially defective ATM/p53 transcriptional and apoptotic responses to various DNA-damaging regimens. In contrast, NOTCH1 or K/N-RAS mutated CLL displayed normal responses in p53/ATM target gene induction and apoptosis. In sole SF3B1 mutated cases, ATM kinase function remained intact, and γH2AX formation, a marker for DNA damage, was increased at baseline and upon irradiation. Our data demonstrate that single mutations in sf3b1 are associated with increased DNA damage and/or an aberrant response to DNA damage. Together, our observations may offer an explanation for the poor prognosis associated with SF3B1 mutations.


European Journal of Immunology | 2004

Apoptosis via the B cell antigen receptor requires Bax translocation and involves mitochondrial depolarization, cytochrome C release, and caspase‐9 activation

Eric Eldering; Wendelina J. M. Mackus; Ingrid A. M. Derks; Ludo M. Evers; Esther Beuling; Peter Teeling; Susanne M. A. Lens; Marinus H. J. van Oers; René A. W. van Lier

Various routes to apoptosis can be active during B cell development. In a model system of mature B cells, differences in caspase‐3 processing have suggested that antigen receptor (BCR)‐mediated apoptosis may involve a zVAD‐insensitive initiator protease(s). In search of the events leading to caspase‐3 activation, we now establish that both CD95‐ and BCR‐mediated apoptosis depend on Bax activation and cytochrome C (cytC) release. Nevertheless, the timing and caspase‐dependence of mitochondrial membrane depolarization differed considerably after CD95‐ or BCR‐triggering. To delineate events subsequent to cytC release, we compared apoptosis induced via BCR triggering and via direct mitochondrial depolarization by CCCP. In both cases, partial processing of caspase‐3 was observed in the presence of zVAD. By expression in 293 cells we addressed the potential of candidate initiator caspases to function in the presence of zVAD, and found that caspase‐9 efficiently processed caspase‐3, while caspase‐2 or –8 were inactive. Finally, retroviral expression of dominant‐negative caspase‐9 inhibited both CD95‐ and BCR‐mediated apoptosis. In conclusion, we obtained no evidence for involvement of a BCR‐specific protease. Instead, our data show for the first time that the BCR‐signal causes Bax translocation, followed by mitochondrial depolarization, and cytC release. Subsequent caspase‐9 activation can solely account for events further downstream.


Blood | 2012

BH3-only protein Noxa regulates apoptosis in activated B cells and controls high-affinity antibody formation

Felix M. Wensveen; Ingrid A. M. Derks; Klaas P. J. M. van Gisbergen; Alex M. de Bruin; Joost C. M. Meijers; HaciAli Yigittop; Martijn A. Nolte; Eric Eldering; René A. W. van Lier

The efficiency of humoral immune responses depends on the selective outgrowth of B cells and plasma cells that produce high affinity antibodies. The factors responsible for affinity maturation of B cell clones in the germinal center (GC) have been well established but selection mechanisms that allow clones to enter the GC are largely unknown. Here we identify apoptosis, regulated by the proapoptotic BH3-only member Noxa (Pmaip1), as a critical factor for the selection of high-affinity clones during B cell expansion after antigen triggering. Noxa is induced in activated B cells, and its ablation provides a survival advantage both in vitro and in vivo. After immunization or influenza infection, Noxa(-/-) mice display enlarged GCs, in which B cells with reduced antigen affinity accumulate. As a consequence, Noxa(-/-) mice mount low affinity antibody responses compared with wild-type animals. Importantly, the low affinity responses correlate with increased immunoglobulin diversity, and cannot be corrected by booster immunization. Thus, normal elimination of low affinity cells favors outgrowth of the remaining high-affinity clones, and this is mandatory for the generation of proper antibody responses. Manipulation of this process may alter the breadth of antibody responses after immunization.

Collaboration


Dive into the Ingrid A. M. Derks'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

E Eldering

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Veronika Navrkalová

Central European Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge