Network


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

Hotspot


Dive into the research topics where Sonia Schoonbroodt is active.

Publication


Featured researches published by Sonia Schoonbroodt.


Biochemical Pharmacology | 2000

Oxidative stress interference with the nuclear factor- κB activation pathways

Sonia Schoonbroodt; Jacques Piette

Abstract While intracellular redox balance is tightly controlled in many cell types, its modification leads to important cellular changes derived, in part, from a modification of the pattern of gene expression. This modification relies on many transcription factors whose activities are either increased or reduced by a disbalance of the redox environment. Among these transcription factors, nuclear factor-κB (NF-κB) plays a pivotal role in inducing genes involved in the control of the immune system as well as in the response to injury and infection. Because NF-κB can be induced in many cells by a diverse set of stimulating agents, it has been proposed that agents activating it do so by increasing oxidative stress within the cell. However, this model was not found to be universal, since the dependence between NF-κB activation and intracellular reactive oxygen species (ROS) generation was only detected in certain cell lines. The origin of this dependency is still unknown, but could very well be situated in a particular kinase or in adaptator molecules of the signaling cascade, leading to inhibitor κBα (IκBα) phosphorylation. On the other hand, NF-κB can be activated by oxidants in many cell types, but this activation is well characterized only in lymphocytes. This activation is distinct from that of classical activators such as proinflammatory cytokines and phorbol esters, because the activation mechanisms appear to converge on a particular tyrosine residue of IκB-α instead of the two classical N-terminal serines. The nature of the protein kinases or protein phosphatases involved in this process is still undetermined. It will be a challenge in the future to identify the kinases/phosphatases activated by oxidants and to discover why ROS are required in some cells to turn on the transduction pathway leading to NF-κB activation by physiological stimuli.


Molecular and Cellular Biology | 1999

Reactive oxygen intermediate-dependent NF-kappaB activation by interleukin-1beta requires 5-lipoxygenase or NADPH oxidase activity.

Giuseppina Bonizzi; Jacques Piette; Sonia Schoonbroodt; Roland Greimers; Laurence Havard; Marie Paule Merville; Vincent Bours

ABSTRACT We previously reported that the role of reactive oxygen intermediates (ROIs) in NF-κB activation by proinflammatory cytokines was cell specific. However, the sources for ROIs in various cell types are yet to be determined and might include 5-lipoxygenase (5-LOX) and NADPH oxidase. 5-LOX and 5-LOX activating protein (FLAP) are coexpressed in lymphoid cells but not in monocytic or epithelial cells. Stimulation of lymphoid cells with interleukin-1β (IL-1β) led to ROI production and NF-κB activation, which could both be blocked by antioxidants or FLAP inhibitors, confirming that 5-LOX was the source of ROIs and was required for NF-κB activation in these cells. IL-1β stimulation of epithelial cells did not generate any ROIs and NF-κB induction was not influenced by 5-LOX inhibitors. However, reintroduction of a functional 5-LOX system in these cells allowed ROI production and 5-LOX-dependent NF-κB activation. In monocytic cells, IL-1β treatment led to a production of ROIs which is independent of the 5-LOX enzyme but requires the NADPH oxidase activity. This pathway involves the Rac1 and Cdc42 GTPases, two enzymes which are not required for NF-κB activation by IL-1β in epithelial cells. In conclusion, three different cell-specific pathways lead to NF-κB activation by IL-1β: a pathway dependent on ROI production by 5-LOX in lymphoid cells, an ROI- and 5-LOX-independent pathway in epithelial cells, and a pathway requiring ROI production by NADPH oxidase in monocytic cells.


Journal of Immunology | 2000

Crucial role of the amino-terminal tyrosine residue 42 and the carboxyl-terminal PEST domain of I kappa B alpha in NF-kappa B activation by an oxidative stress.

Sonia Schoonbroodt; Valérie Ferreira; Martin Best-Belpomme; Johan R. Boelaert; Sylvie Legrand-Poels; Marie Körner; Jacques Piette

Activation of transcription factor NF-κB involves the signal-dependent degradation of basally phosphorylated inhibitors such as IκBα. In response to proinflammatory cytokines or mitogens, the transduction machinery has recently been characterized, but the activation mechanism upon oxidative stress remains unknown. In the present work, we provide several lines of evidence that NF-κB activation in a T lymphocytic cell line (EL4) by hydrogen peroxide (H2O2) did not involve phosphorylation of the serine residues 32 and 36 in the amino-terminal part of IκBα. Indeed, mutation of Ser32 and Ser36 blocked IL-1β- or PMA-induced NF-κB activation, but had no effect on its activation by H2O2. Although IκBα was phosphorylated upon exposure to H2O2, tyrosine residue 42 and the C-terminal PEST (proline-glutamic acid-serine-threonine) domain played an important role. Indeed, mutation of tyrosine 42 or serine/threonine residues of the PEST domain abolished NF-κB activation by H2O2, while it had no effect on activation by IL-1β or PMA-ionomycin. This H2O2-inducible phosphorylation was not dependent on IκB kinase activation, but could involve casein kinase II, because an inhibitor of this enzyme (5,6-dichloro-1-β-d-ribofuranosyl-benzimidazole) blocks NF-κB activation. H2O2-induced IκBα phosphorylation was followed by its degradation by calpain proteases or through the proteasome. Taken together, our findings suggest that NF-κB activation by H2O2 involves a new mechanism that is totally distinct from those triggered by proinflammatory cytokines or mitogens.


Oncogene | 1999

The ATM protein is required for sustained activation of NF-κB following DNA damage

Bernard Piret; Sonia Schoonbroodt; Jacques Piette

Cells lacking an intact ATM gene are hypersensitive to ionizing radiation and show multiple defects in the cell cycle-coupled checkpoints. DNA damage usually triggers cell cycle arrest through, among other things, the activation of p53. Another DNA-damage responsive factor is NF-κB. It is activated by various stress situations, including oxidative stress, and by DNA-damaging compounds such as topoisomerase poisons. We found that cells from Ataxia Telangiectasia patients exhibit a defect in NF-κB activation in response to treatment with camptothecin, a topoisomerase I poison. In AT cells, this activation is shortened or suppressed, compared to that observed in normal cells. Ectopic expression of the ATM protein in AT cells increases the activation of NF-κB in response to camptothecin. MO59J glioblastoma cells that do not express the DNA-PK catalytic subunit respond normally to camptothecin. These results support the hypothesis that NF-κB is a DNA damage-responsive transcription factor and that its activation pathway by DNA damage shares some components with the one leading to p53 activation.


Journal of Photochemistry and Photobiology B-biology | 1998

NF-κB: an important transcription factor in photobiology

Sylvie Legrand-Poels; Sonia Schoonbroodt; Jean-Yves Matroule; Jacques Piette

Increased gene expression as a consequence of environmental stress is typically observed in mammalian cells. In the past few years the cis- and trans-acting genetic elements responsible for gene induction by radiation (from UV-C to visible light) started to be well characterized. The molecular mechanisms involved in the cell response to radiation reveal that an important control occurs at the transcriptional level and is coordinated by various transcription factors. Among these transcription factors, the well-known Rel/NF-kappa B family of vertebrate transcription factors plays a pivotal role as it controls both the inflammatory and immune responses. The NF-kappa B family comprises a number of structurally related, interacting proteins that bind DNA as dimers and whose activity is regulated by subcellular location. This family includes many members (p50, p52, RelA, RelB, c-Rel, ...), most of which can form DNA-binding homo- or heterodimers. Nuclear expression and consequent biological action of the eukaryotic NF-kappa B transcription factor complex are tightly regulated through its cytoplasmic retention by ankyrin-rich inhibitory proteins known as I kappa B. In the best-characterized example, I kappa B-alpha interacts with a p50/RelA (NF-kappa B) heterodimer to retain the complex in the cytoplasm and inhibit its DNA-binding activity. Upon receiving a variety of signals, many of which are probably mediated by the generation of reactive oxygen species (ROS), I kappa B-alpha undergoes phosphorylation, is then ubiquitinated at nearby lysine residues and finally degraded by the proteasome, while still complexed with NF- kappa B. Removal of I kappa B-alpha uncovers the nuclear localization signals on subunits of NF-kappa B, allowing the complex to enter the nucleus, bind to DNA and affect gene expression. In this paper, we shall show that molecular mechanisms leading to NF-kappa B activation by UV or by photosensitization are initiated by oxidative damage at the membrane level or by the induction of DNA alterations. While the exact nature of the transduction intermediates is still unknown, we shall show that NF-kappa B activation by radiation follows different pathways from those used by pro-inflammatory cytokines.


Journal of General Virology | 1995

Varicella-zoster virus induces apoptosis in cell culture.

Catherine Sadzot-Delvaux; Pascale Thonard; Sonia Schoonbroodt; Jacques Piette; Bernard Rentier

Apoptosis is an active mechanism of cell death which can be initiated in response to various stimuli including virus infections. In this work, we demonstrate that lytic infection by varicella-zoster virus (VZV), a human herpesvirus, is characterized by nuclear fragmentation of DNA into oligonucleosomal fragments and by chromatin condensation. In vitro, VZV-induced cell death is actually mediated by apoptosis. The mechanisms developed by cells to protect themselves against apoptosis could be one of the parameters allowing the establishment of virus latency. In the case of VZV, which can remain latent in sensory ganglia, we have not yet identified a cellular or viral protein which could play this protective role, since the observed apoptosis mechanism seems to be independent from Bcl-2, the most frequently described inhibitor of apoptosis.


Journal of Immunology | 2008

Engineering Antibody Heavy Chain CDR3 to Create a Phage Display Fab Library Rich in Antibodies That Bind Charged Carbohydrates

Sonia Schoonbroodt; Mieke Steukers; Malini Viswanathan; Nicolas Frans; Marie Timmermans; Anita Wehnert; Minh Nguyen; Robert Charles Ladner; Rene Hoet

A number of small charged carbohydrate moieties have been associated with inflammation and cancer. However, the development of therapeutic Abs targeting these moieties has been hampered by their low immunogenicity and their structural relationship to self-Ag. We report the design of an Ab repertoire enriched in Abs binding to small charged carbohydrates and the construction of a human Fab phagemid library, “FAB-CCHO.” This library combines L chain Ig sequences from human donors and H chain synthetic diversity constructed in key Ag contact sites in CDRs 1, 2, and 3 of the human framework VH3–23. The H chain CDR3 has been engineered to enrich the library in Abs that bind charged carbohydrates by the introduction of basic residues at specific amino acid locations. These residues were selected on the basis of anti-carbohydrate Ab sequence alignment. The success of this design is demonstrated by the isolation of phage Abs against charged carbohydrate therapeutic target Ags such as sulfated sialyl-Lewis X glycan and heparan sulfate.


Biochemical Pharmacology | 1999

Role of the protein kinase C λ/ι isoform in nuclear factor-κB activation by interleukin-1β or tumor necrosis factor-α: cell type specificities

Giuseppina Bonizzi; Jacques Piette; Sonia Schoonbroodt; Marie Paule Merville; Vincent Bours

Abstract It has previously been reported that distinct signaling pathways can lead to nuclear factor (NF)-κB activation following stimulation of different cell types with inflammatory cytokines. As the role of atypical protein kinase C (PKC) isoforms in NF-κB activation remains a matter of controversy, we investigated whether this role might be cell type-dependent. Immunoblots detected atypical PKC expression in all the analyzed cell lines. The PKC inhibitor calphostin C inhibited NF-κB activation by tumor necrosis factor (TNF)-α or interleukin (IL)-1β in Jurkat or NIH3T3 cells but not in MCF7 A/Z cells. Cell transfections with a PKC λ/ι dominant negative mutant abolished TNF-α-induced NF-κB-dependent transcription in NIH3T3 and Jurkat cells but not in MCF7 A/Z cells. Similarly, the same mutant blocked NF-κB-dependent transactivation after IL-1β stimulation of NIH3T3 cells, but was ineffective after IL-1β treatment of MCF7 A/Z cells. In MCF7 A/Z cells, however, the PKC λ/ι dominant negative mutant could abolish transactivation of an AP-1-dependent reporter plasmid after stimulation with TNF-α but not with IL-1β. These data thus confirm that transduction pathways for NF-κB activation after cell stimulation with TNF-α or IL-1β are cell-type specific and that atypical PKC isoforms participate in this pathway in NIH3T3 and Jurkat cells.


Free Radical Research | 1997

Involvement of different transduction pathways in NF-kappaB activation by several inducers

Sylvie Legrand-Poels; Laurent Zecchinon; Bernard Piret; Sonia Schoonbroodt; Jacques Piette

Double-stimulation was used to demonstrate that, in a T lymphocytic cell line (CEM), phorbol myristate acetate (PMA) rapidly induced NF-kappa B through a signaling pathway which did not involve reactive oxygen species (ROS) and was different from the activation triggered by either H2O2 or tumor necrosis factor-alpha (TNF-alpha). Since these latter compounds were known to activate NF-kappa B translocation in a redox-sensitive way, we have demonstrated that NF-kappa B activation by PMA was resistant to antioxidant N-acetyl-L-cysteine (NAC) and sensitive to kinase inhibitors staurosporine and H7 while activation by H2O2 or TNF-alpha were not.


Molecular and Cellular Biology | 2003

Potentiation of Tumor Necrosis Factor-Induced NF-κB Activation by Deacetylase Inhibitors Is Associated with a Delayed Cytoplasmic Reappearance of IκBα

Emmanuelle Adam; Vincent Quivy; Françoise Bex; Alain Chariot; Yves Collette; Caroline Vanhulle; Sonia Schoonbroodt; Véronique Goffin; Thi Lien-Anh Nguyen; Geoffrey Gloire; Géraldine Carrard; Bertrand Friguet; Yvan de Launoit; Arsène Burny; Vincent Bours; Jacques Piette; Carine Van Lint

Previous studies have implicated acetylases and deacetylases in regulating the transcriptional activity of NF-B. Here, we show that inhibitors of deacetylases such as trichostatin A (TSA) and sodium butyrate (NaBut) potentiated TNF-induced expression of several natural NF-B-driven promoters. This transcriptional synergism observed between TNF and TSA (or NaBut) required intact B sites in all promoters tested and was biologically relevant as demonstrated by RNase protection on two instances of endogenous NF-B-regulated gene transcription. Importantly, TSA prolonged both TNF-induced DNA-binding activity and the presence of NF-B in the nucleus. We showed that the p65 subunit of NF-B was acetylated in vivo. However, this acetylation was weak, suggesting that other mechanisms could be implicated in the potentiated binding and transactivation activities of NF-B after TNF plus TSA versus TNF treatment. Western blot and immunofluorescence confocal microscopy experiments revealed a delay in the cytoplasmic reappearance of the IB inhibitor that correlated temporally with the prolonged intranuclear binding and presence of NF-B. This delay was due neither to a defect in IB mRNA production nor to a nuclear retention of IB but was rather due to a persistent proteasome-mediated degradation of IB. A prolongation of IB kinase activity could explain, at least partially, the delayed IB cytoplasmic reappearance observed in presence of TNF plus TSA.

Collaboration


Dive into the Sonia Schoonbroodt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rene Hoet

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carine Van Lint

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emmanuelle Adam

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge