Network


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

Hotspot


Dive into the research topics where Filip Van Hauwermeiren is active.

Publication


Featured researches published by Filip Van Hauwermeiren.


Immunity | 2011

RIP kinase-dependent necrosis drives lethal systemic inflammatory response syndrome.

Linde Duprez; Nozomi Takahashi; Filip Van Hauwermeiren; Benjamin Vandendriessche; Vera Goossens; Tom Vanden Berghe; Wim Declercq; Claude Libert; Anje Cauwels; Peter Vandenabeele

Engagement of tumor necrosis factor receptor 1 signals two diametrically opposed pathways: survival-inflammation and cell death. An additional switch decides, depending on the cellular context, between caspase-dependent apoptosis and RIP kinase (RIPK)-mediated necrosis, also termed necroptosis. We explored the contribution of both cell death pathways in TNF-induced systemic inflammatory response syndrome (SIRS). Deletion of apoptotic executioner caspases (caspase-3 or -7) or inflammatory caspase-1 had no impact on lethal SIRS. However, deletion of RIPK3 conferred complete protection against lethal SIRS and reduced the amounts of circulating damage-associated molecular patterns. Pretreatment with the RIPK1 kinase inhibitor, necrostatin-1, provided a similar effect. These results suggest that RIPK1-RIPK3-mediated cellular damage by necrosis drives mortality during TNF-induced SIRS. RIPK3 deficiency also protected against cecal ligation and puncture, underscoring the clinical relevance of RIPK kinase inhibition in sepsis and identifying components of the necroptotic pathway that are potential therapeutic targets for treatment of SIRS and sepsis.


Cytokine & Growth Factor Reviews | 2011

Treatment of TNF mediated diseases by selective inhibition of soluble TNF or TNFR1

Filip Van Hauwermeiren; Roosmarijn E. Vandenbroucke; Claude Libert

The TNF signaling pathway is a valuable target in the therapy of autoimmune diseases, and anti-TNF drugs are successfully used to treat diseases such as rheumatoid arthritis, Crohns disease and psoriasis. By their ability to interfere with inflammatory processes at multiple levels, these TNF blockers have become invaluable tools to inhibit the inflammation induced damage and allow recovery of the affected tissues. Unfortunately this therapy has some drawbacks, including increased risk of infection and malignancy, and remarkably, the onset of new auto-immune diseases. Some of these effects are caused by the unwanted abrogation of beneficial TNF signaling. More specific targeting of the pathological TNF-induced signaling might lead to broader applicability and improved safety. Specificity might be increased by inhibiting the soluble TNF/TNFR1 axis while leaving the often beneficial transmembrane TNF/TNFR2 signaling untouched. This approach looks promising because it inhibits the pathological effects of TNF and reduces the side effects, and it opens the way for the treatment of other diseases in which TNFR2 inhibition is detrimental. In this review we give an overview of in vivo mouse studies of TNF mediated pathologies demonstrating that the blockade or genetic deletion of sTNF or TNFR1 is preferable over total TNF blockade.


The Journal of Neuroscience | 2012

Matrix Metalloprotease 8-Dependent Extracellular Matrix Cleavage at the Blood–CSF Barrier Contributes to Lethality during Systemic Inflammatory Diseases

Roosmarijn E. Vandenbroucke; Eline Dejonckheere; Philippe Van Lint; Delphine Demeestere; Elien Van Wonterghem; Ineke Vanlaere; Leen Puimège; Filip Van Hauwermeiren; Riet De Rycke; Conor Mc Guire; Cristina Campestre; Carlos López-Otín; Patrick Matthys; Georges Leclercq; Claude Libert

Systemic inflammatory response syndrome (SIRS) is a highly mortal inflammatory disease, associated with systemic inflammation and organ dysfunction. SIRS can have a sterile cause or can be initiated by an infection, called sepsis. The prevalence is high, and available treatments are ineffective and mainly supportive. Consequently, there is an urgent need for new treatments. The brain is one of the first organs affected during SIRS, and sepsis and the consequent neurological complications, such as encephalopathy, are correlated with decreased survival. The choroid plexus (CP) that forms the blood–CSF barrier (BCSFB) is thought to act as a brain “immune sensor” involved in the communication between the peripheral immune system and the CNS. Nevertheless, the involvement of BCSFB integrity in systemic inflammatory diseases is seldom investigated. We report that matrix metalloprotease-8 (MMP8) depletion or inhibition protects mice from death and hypothermia in sepsis and renal ischemia/reperfusion. This effect could be attributed to MMP8-dependent leakage of the BCSFB, caused by collagen cleavage in the extracellular matrix of CP cells, which leads to a dramatic change in cellular morphology. Disruption of the BCSFB results in increased CSF cytokine levels, brain inflammation, and downregulation of the brain glucocorticoid receptor. This receptor is necessary for dampening the inflammatory response. Consequently, MMP8+/+ mice, in contrast to MMP8−/− mice, show no anti-inflammatory response and this results in high mortality. In conclusion, we identify MMP8 as an essential mediator in SIRS and, hence, a potential drug target. We also propose that the mechanism of action of MMP8 involves disruption of the BCSFB integrity.


Journal of Experimental Medicine | 2009

Type I interferon drives tumor necrosis factor–induced lethal shock

Liesbeth Huys; Filip Van Hauwermeiren; Lien Dejager; Eline Dejonckheere; Stefan Lienenklaus; Siegfried Weiss; Georges Leclercq; Claude Libert

Tumor necrosis factor (TNF) is reputed to have very powerful antitumor effects, but it is also a strong proinflammatory cytokine. Injection of TNF in humans and mice leads to a systemic inflammatory response syndrome with major effects on liver and bowels. TNF is also a central mediator in several inflammatory diseases. We report that type I interferons (IFNs) are essential mediators of the lethal response to TNF. Mice deficient in the IFN-α receptor 1 (IFNAR-1) or in IFN-β are remarkably resistant to TNF-induced hypothermia and death. After TNF injection, IFNAR-1−/− mice produced less IL-6, had less bowel damage, and had less apoptosis of enterocytes and hepatocytes compared with wild-type (WT) mice. Extensive gene expression analysis in livers of WT and IFNAR-1−/− mice revealed a large deficiency in the response to TNF in the knockout mice, especially of IFN-stimulated response element–dependent genes, many of which encode chemokines. In livers of IFNAR-1−/− mice, fewer infiltrating white blood cells (WBCs) were detected by immunohistochemistry. Deficiency of type I IFN signaling provided sufficient protection for potentially safer therapeutic use of TNF in tumor-bearing mice. Our data illustrate that type I IFNs act as essential mediators in TNF-induced lethal inflammatory shock, possibly by enhancing cell death and inducing chemokines and WBC infiltration in tissues.


Embo Molecular Medicine | 2013

Matrix metalloproteinase 13 modulates intestinal epithelial barrier integrity in inflammatory diseases by activating TNF.

Roosmarijn E. Vandenbroucke; Eline Dejonckheere; Filip Van Hauwermeiren; Sofie Lodens; Riet De Rycke; Elien Van Wonterghem; An Staes; Kris Gevaert; Carlos López-Otín; Claude Libert

Several pathological processes, such as sepsis and inflammatory bowel disease (IBD), are associated with impairment of intestinal epithelial barrier. Here, we investigated the role of matrix metalloproteinase MMP13 in these diseases. We observed that MMP13−/− mice display a strong protection in LPS‐ and caecal ligation and puncture‐induced sepsis. We could attribute this protection to reduced LPS‐induced goblet cell depletion, endoplasmic reticulum stress, permeability and tight junction destabilization in the gut of MMP13−/− mice compared to MMP13+/+ mice. Both in vitro and in vivo, we found that MMP13 is able to cleave pro‐TNF into bioactive TNF. By LC‐MS/MS, we identified three MMP13 cleavage sites, which proves that MMP13 is an alternative TNF sheddase next to the TNF converting enzyme TACE. Similarly, we found that the same mechanism was responsible for the observed protection of the MMP13−/− mice in a mouse model of DSS‐induced colitis. We identified MMP13 as an important mediator in sepsis and IBD via the shedding of TNF. Hence, we propose MMP13 as a novel drug target for diseases in which damage to the gut is essential.


Journal of Clinical Investigation | 2013

Safe TNF-based antitumor therapy following p55TNFR reduction in intestinal epithelium

Filip Van Hauwermeiren; Marietta Armaka; Niki Karagianni; Ksanthi Kranidioti; Roosmarijn E. Vandenbroucke; Sonja Loges; Maarten Van Roy; Jan Staelens; Leen Puimège; Ajay Palagani; Wim Vanden Berghe; Panayiotis Victoratos; Peter Carmeliet; Claude Libert; George Kollias

TNF has remarkable antitumor activities; however, therapeutic applications have not been possible because of the systemic and lethal proinflammatory effects induced by TNF. Both the antitumor and inflammatory effects of TNF are mediated by the TNF receptor p55 (p55TNFR) (encoded by the Tnfrsf1a gene). The antitumor effect stems from an induction of cell death in tumor endothelium, but the cell type that initiates the lethal inflammatory cascade has been unclear. Using conditional Tnfrsf1a knockout or reactivation mice, we found that the expression level of p55TNFR in intestinal epithelial cells (IECs) is a crucial determinant in TNF-induced lethal inflammation. Remarkably, tumor endothelium and IECs exhibited differential sensitivities to TNF when p55TNFR levels were reduced. Tumor-bearing Tnfrsf1a⁺⁺/⁻ or IEC-specific p55TNFR-deficient mice showed resistance to TNF-induced lethality, while the tumor endothelium remained fully responsive to TNF-induced apoptosis and tumors regressed. We demonstrate proof of principle for clinical application of this approach using neutralizing anti-human p55TNFR antibodies in human TNFRSF1A knockin mice. Our results uncover an important cellular basis of TNF toxicity and reveal that IEC-specific or systemic reduction of p55TNFR mitigates TNF toxicity without loss of antitumor efficacy.


Journal of Biological Chemistry | 2011

Tumor Necrosis Factor Inhibits Glucocorticoid Receptor Function in Mice A STRONG SIGNAL TOWARD LETHAL SHOCK

Tom Van Bogaert; Sofie Vandevyver; Lien Dejager; Filip Van Hauwermeiren; Iris Pinheiro; Ioanna Petta; David Engblom; Anna Kleyman; Guenther Schutz; Jan Tuckermann; Claude Libert

As glucocorticoid resistance (GCR) and the concomitant burden pose a worldwide problem, there is an urgent need for a more effective glucocorticoid therapy, for which insights into the molecular mechanisms of GCR are essential. In this study, we addressed the hypothesis that TNFα, a strong pro-inflammatory mediator in numerous inflammatory diseases, compromises the protective function of the glucocorticoid receptor (GR) against TNFα-induced lethal inflammation. Indeed, protection of mice by dexamethasone against TNFα lethality was completely abolished when it was administered after TNFα stimulation, indicating compromised GR function upon TNFα challenge. TNFα-induced GCR was further demonstrated by impaired GR-dependent gene expression in the liver. Furthermore, TNFα down-regulates the levels of both GR mRNA and protein. However, this down-regulation seems to occur independently of GC production, as TNFα also resulted in down-regulation of GR levels in adrenalectomized mice. These findings suggest that the decreased amount of GR determines the GR response and outcome of TNFα-induced shock, as supported by our studies with GR heterozygous mice. We propose that by inducing GCR, TNFα inhibits a major brake on inflammation and thereby amplifies the pro-inflammatory response. Our findings might prove helpful in understanding GCR in inflammatory diseases in which TNFα is intimately involved.


Immunity | 2017

Nlrp6- and ASC-Dependent Inflammasomes Do Not Shape the Commensal Gut Microbiota Composition

Michail Mamantopoulos; Francesca Ronchi; Filip Van Hauwermeiren; Sara Vieira-Silva; Bahtiyar Yilmaz; Liesbet Martens; Yvan Saeys; Stefan K Drexler; Amir S. Yazdi; Jeroen Raes; Mohamed Lamkanfi; Kathleen McCoy; Andy Wullaert

&NA; The gut microbiota regulate susceptibility to multiple human diseases. The Nlrp6‐ASC inflammasome is widely regarded as a hallmark host innate immune axis that shapes the gut microbiota composition. This notion stems from studies reporting dysbiosis in mice lacking these inflammasome components when compared with non‐littermate wild‐type animals. Here, we describe microbial analyses in inflammasome‐deficient mice while minimizing non‐genetic confounders using littermate‐controlled Nlrp6‐deficient mice and ex‐germ‐free littermate‐controlled ASC‐deficient mice that were all allowed to shape their gut microbiota naturally after birth. Careful microbial phylogenetic analyses of these cohorts failed to reveal regulation of the gut microbiota composition by the Nlrp6‐ and ASC‐dependent inflammasomes. Our results obtained in two geographically separated animal facilities dismiss a generalizable impact of Nlrp6‐ and ASC‐dependent inflammasomes on the composition of the commensal gut microbiota and highlight the necessity for littermate‐controlled experimental design in assessing the influence of host immunity on gut microbial ecology. HighlightsNon‐littermate controls fail to define host genetic impacts on the gut microbiotaLittermate separation does not reveal ASC‐Nlrp6 impacts on the gut microbiotaLifetime littermate separation does not reveal Nlrp6 impacts on DSS colitis &NA; Inflammasomes were proposed to shape gut ecology based on dysbiosis in mutant mice versus non‐littermate wild‐types. Mamantopoulos et al. show that inflammasomes do not affect gut microbiota composition when controlling for non‐genetic confounders. This finding dismisses the suggested role for inflammasomes in controlling host health through regulation of intestinal ecology.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Familial Mediterranean fever mutations lift the obligatory requirement for microtubules in Pyrin inflammasome activation

Hanne Van Gorp; Pedro Henrique Viana Saavedra; Nathalia Moraes de Vasconcelos; Nina Van Opdenbosch; Lieselotte Vande Walle; Magdalena Matusiak; Giusi Prencipe; Antonella Insalaco; Filip Van Hauwermeiren; Dieter Demon; Delfien Bogaert; Melissa Dullaers; Elfride De Baere; Tino Hochepied; Jo Dehoorne; Karim Vermaelen; Filomeen Haerynck; Fabrizio De Benedetti; Mohamed Lamkanfi

Significance Familial Mediterranean fever (FMF) is an autoinflammatory disease caused by more than 310 mutations in the gene MEFV, which encodes Pyrin. Pyrin recently was shown to trigger inflammasome activation in response to Rho GTPase-modifying bacterial toxins. Here we report that Clostridium difficile infection and intoxication with its enterotoxin TcdA engage the Pyrin inflammasome. Moreover, activation of the Pyrin inflammasome, but not other inflammasomes, was hampered by microtubule-depolymerizing drugs in mouse and humans. Unexpectedly, we found that FMF mutations render Pyrin activation independent of microtubules. Thus, our findings provide a conceptual framework for understanding Pyrin signaling and enable functional diagnosis of FMF. Familial Mediterranean fever (FMF) is the most common monogenic autoinflammatory disease worldwide. It is caused by mutations in the inflammasome adaptor Pyrin, but how FMF mutations alter signaling in FMF patients is unknown. Herein, we establish Clostridium difficile and its enterotoxin A (TcdA) as Pyrin-activating agents and show that wild-type and FMF Pyrin are differentially controlled by microtubules. Diverse microtubule assembly inhibitors prevented Pyrin-mediated caspase-1 activation and secretion of IL-1β and IL-18 from mouse macrophages and human peripheral blood mononuclear cells (PBMCs). Remarkably, Pyrin inflammasome activation persisted upon microtubule disassembly in PBMCs of FMF patients but not in cells of patients afflicted with other autoinflammatory diseases. We further demonstrate that microtubules control Pyrin activation downstream of Pyrin dephosphorylation and that FMF mutations enable microtubule-independent assembly of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) micrometer-sized perinuclear structures (specks). The discovery that Pyrin mutations remove the obligatory requirement for microtubules in inflammasome activation provides a conceptual framework for understanding FMF and enables immunological screening of FMF mutations.


Journal of Biological Chemistry | 2015

Generation and Characterization of Small Single Domain Antibodies Inhibiting Human Tumor Necrosis Factor Receptor 1

Sophie Steeland; Leen Puimège; Roosmarijn E. Vandenbroucke; Filip Van Hauwermeiren; Jurgen Haustraete; Nick Devoogdt; Paco Hulpiau; Geert Leroux-Roels; Debby Laukens; Philip Meuleman; Martine De Vos; Claude Libert

Background: Several anti-TNF biologicals are available to treat autoimmune diseases. However, selective TNFR1 inhibition is advisable, thereby reducing the pro-inflammatory TNF/TNFR1 signaling, while the good immunomodulatory TNF/TNFR2 signaling is preserved. Results: We generated and characterized an anti-TNFR1 Nanobody, TNF Receptor-One Silencer (TROS). Conclusion: TROS inhibits inflammation in vitro, ex vivo, and in vivo. Significance: Anti-TNFR1 therapies are potential novel treatments against autoimmune diseases. The cytokine TNF is a well known drug target for several inflammatory diseases such as Crohn disease. Despite the great success of TNF blockers, therapy could be improved because of high costs and side effects. Selective inhibition of TNF receptor (TNFR) 1 signaling holds the potential to greatly reduce the pro-inflammatory activity of TNF, thereby preserving the advantageous immunomodulatory signals mediated by TNFR2. We generated a selective human TNFR1 inhibitor based on Nanobody (Nb) technology. Two anti-human TNFR1 Nbs were linked with an anti-albumin Nb to generate Nb Alb-70-96 named “TNF Receptor-One Silencer” (TROS). TROS selectively binds and inhibits TNF/TNFR1 and lymphotoxin-α/TNFR1 signaling with good affinity and IC50 values, both of which are in the nanomolar range. Surface plasmon resonance analysis reveals that TROS competes with TNF for binding to human TNFR1. In HEK293T cells, TROS strongly reduces TNF-induced gene expression, like IL8 and TNF, in a dose-dependent manner; and in ex vivo cultured colon biopsies of CD patients, TROS inhibits inflammation. Finally, in liver chimeric humanized mice, TROS antagonizes inflammation in a model of acute TNF-induced liver inflammation, reflected in reduced human IL8 expression in liver and reduced IL6 levels in serum. These results demonstrate the considerable potential of TROS and justify the evaluation of TROS in relevant disease animal models of both acute and chronic inflammation and eventually in patients.

Collaboration


Dive into the Filip Van Hauwermeiren'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge