Network


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

Hotspot


Dive into the research topics where J. Sjef Verbeek is active.

Publication


Featured researches published by J. Sjef Verbeek.


Immunity | 2002

Arthritis Critically Dependent on Innate Immune System Players

Hong Ji; Koichiro Ohmura; Umar Mahmood; David M. Lee; Frans M. A. Hofhuis; Susan A. Boackle; Kazue Takahashi; V. Michael Holers; Mark Walport; Craig Gerard; Alan Ezekowitz; Michael C. Carroll; Michael B. Brenner; Ralph Weissleder; J. Sjef Verbeek; Véronique Duchatelle; Claude Degott; Christophe Benoist; Diane Mathis

K/BxN T cell receptor transgenic mice are a model of inflammatory arthritis, similar to rheumatoid arthritis. Disease in these animals is focused specifically on the joints but stems from autoreactivity to a ubiquitously expressed antigen, glucose-6-phosphate isomerase (GPI). T and B cells are both required for disease initiation, but anti-GPI immunoglobulins (Igs), alone, can induce arthritis in lymphocyte-deficient recipients. Here, we show that the arthritogenic Igs act through both Fc receptors (in particular, FcgammaRIII) and the complement network (C5a). Surprisingly, the alternative pathway of complement activation is critical, while classical pathway components are entirely dispensable. We suggest that autoimmune disease, even one that is organ specific, can occur when mobilization of an adaptive immune response results in runaway activation of the innate response.


Immunity | 1996

Impaired IgG-dependent anaphylaxis and Arthus reaction in FcγRIII (CD16) deficient mice.

Wouter L. W. Hazenbos; J. Engelbert Gessner; Frans M. A. Hofhuis; Henri Kuipers; Dirk Meyer; Ingmar A. F. M. Heijnen; Reinhold E. Schmidt; Matyas Sandor; P. J. A. Capel; Marc Daëron; Jan G. J. van de Winkel; J. Sjef Verbeek

Abstract The family of receptors for IgG (FcγR) plays an essential role in antibody-mediated effector functions of the immune system. However, the specific contribution of each of the FcγR classes to in vivo immune reactions is still unclear. Here, we demonstrate that mice deficient for the ligand-binding α chain of FcγRIII lack NK cell–mediated antibody-dependent cytotoxicity and phagocytosis of IgG1-coated particles by macrophages. Strikingly, these mice lack IgG-mediated mast cell degranulation, are resistant to IgG-dependent passive cutaneous anaphylaxis, and exhibit an impaired Arthus reaction. These results indicate a prominent role for FcγRIII in inflammatory and anaphylactic responses, making this receptor a potential target in immunotherapy.


Nature Medicine | 2001

Complement facilitates early prion pathogenesis

Michael A. Klein; Pascal S. Kaeser; Petra Schwarz; Heiko Weyd; Ioannis Xenarios; Rolf M. Zinkernagel; Michael C. Carroll; J. Sjef Verbeek; Marina Botto; Mark Walport; Hector Molina; Ulrich Kalinke; Hans Acha-Orbea; Adriano Aguzzi

New-variant Creutzfeldt–Jakob disease and scrapie are typically initiated by extracerebral exposure to the causative agent, and exhibit early prion replication in lymphoid organs. In mouse scrapie, depletion of B-lymphocytes prevents neuropathogenesis after intraperitoneal inoculation, probably due to impaired lymphotoxin-dependent maturation of follicular dendritic cells (FDCs), which are a major extracerebral prion reservoir. FDCs trap immune complexes with Fc-γ receptors and C3d/C4b-opsonized antigens with CD21/CD35 complement receptors. We examined whether these mechanisms participate in peripheral prion pathogenesis. Depletion of circulating immunoglobulins or of individual Fc-γ receptors had no effect on scrapie pathogenesis if B-cell maturation was unaffected. However, mice deficient in C3, C1q, Bf/C2, combinations thereof or complement receptors were partially or fully protected against spongiform encephalopathy upon intraperitoneal exposure to limiting amounts of prions. Splenic accumulation of prion infectivity and PrPSc was delayed, indicating that activation of specific complement components is involved in the initial trapping of prions in lymphoreticular organs early after infection.


Blood | 2010

Antigenic modulation limits the efficacy of anti-CD20 antibodies: implications for antibody selection

Stephen A. Beers; Ruth R. French; H. T. Claude Chan; Sean H. Lim; Timothy C. Jarrett; Regina Mora Vidal; Sahan S. Wijayaweera; Sandra V. Dixon; Hyungjin Kim; Kerry L. Cox; Jonathan P. Kerr; David A. Johnston; Peter Johnson; J. Sjef Verbeek; Martin J. Glennie; Mark S. Cragg

Rituximab, a monoclonal antibody that targets CD20 on B cells, is now central to the treatment of a variety of malignant and autoimmune disorders. Despite this success, a substantial proportion of B-cell lymphomas are unresponsive or develop resistance, hence more potent anti-CD20 monoclonal antibodies (mAbs) are continuously being sought. Here we demonstrate that type II (tositumomab-like) anti-CD20 mAbs are 5 times more potent than type I (rituximab-like) reagents in depleting human CD20 Tg B cells, despite both operating exclusively via activatory Fcgamma receptor-expressing macrophages. Much of this disparity in performance is attributable to type I mAb-mediated internalization of CD20 by B cells, leading to reduced macrophage recruitment and the degradation of CD20/mAb complexes, shortening mAb half-life. Importantly, human B cells from healthy donors and most cases of chronic lymphatic leukemia and mantle cell lymphoma, showed rapid CD20 internalization that paralleled that seen in the Tg mouse B cells, whereas most follicular lymphoma and diffuse large B-cell lymphoma cells were far more resistant to CD20 loss. We postulate that differences in CD20 modulation may play a central role in determining the relative efficacy of rituximab in treating these diseases and strengthen the case for focusing on type II anti-CD20 mAb in the clinic.


Journal of Immunology | 2002

Antigen-Antibody Immune Complexes Empower Dendritic Cells to Efficiently Prime Specific CD8+ CTL Responses In Vivo

Danita H. Schuurhuis; Andreea Ioan-Facsinay; Bas Nagelkerken; Jolien J. van der Ploeg-van Schip; Christine Sedlik; Cornelis J. M. Melief; J. Sjef Verbeek; Ferry Ossendorp

Dendritic cells (DCs) require a maturation signal to acquire efficient CTL-priming capacity. In vitro FcγR-mediated internalization of Ag-Ab immune complexes (ICs) can induce maturation of DCs. In this study, we show that IC-induced DC maturation in vitro enables DCs to prime peptide-specific CD8+ CTLs in vivo, independently of CD4+ Th cells. Importantly, OVA/anti-OVA IC-treated DCs not only primed CD8+ CTLs to an exogenously loaded peptide nonrelated to OVA, but also efficiently primed CTLs against the dominant CTL epitope derived from the OVA Ag present in the ICs. Our studies show that ICs fulfill a dual role in priming of CD8+ CTL responses to exogenous Ags: enhancement of Ag uptake by DCs and activation of DCs, resulting in “license to kill.” These findings indicate that the presence of specific Abs can crucially affect the induction of cytotoxic cellular responses.


Journal of Clinical Investigation | 2004

Anti-C1q autoantibodies deposit in glomeruli but are only pathogenic in combination with glomerular C1q-containing immune complexes

Leendert A. Trouw; Tom W. L. Groeneveld; Marc A. Seelen; Jacques M.G.J. Duijs; Ingeborg M. Bajema; Frans A. Prins; Uday Kishore; David J. Salant; J. Sjef Verbeek; Cees van Kooten; Mohamed R. Daha

Anti-C1q autoantibodies are present in sera of patients with several autoimmune diseases, including systemic lupus erythematosus (SLE). Strikingly, in SLE the presence of anti-C1q is associated with the occurrence of nephritis. We have generated mouse anti-mouse C1q mAbs and used murine models to investigate whether anti-C1q autoantibodies actually contribute to renal pathology in glomerular immune complex disease. Administration of anti-C1q mAb JL-1, which recognizes the collagen-like region of C1q, resulted in glomerular deposition of C1q and anti-C1q autoantibodies and mild granulocyte influx, but no overt renal damage. However, combination of JL-1 with a subnephritogenic dose of C1q-fixing anti-glomerular basement membrane (anti-GBM) antibodies enhanced renal damage characterized by persistently increased levels of infiltrating granulocytes, major histological changes, and increased albuminuria. This was not observed when a non-C1q-fixing anti-GBM preparation was used. Experiments with different knockout mice showed that renal damage was dependent not only on glomerular C1q and complement activation but also on Fcgamma receptors. In conclusion, anti-C1q autoantibodies deposit in glomeruli together with C1q but induce overt renal disease only in the context of glomerular immune complex disease. This provides an explanation why anti-C1q antibodies are especially pathogenic in patients with SLE.


Journal of Experimental Medicine | 2002

Complement Activation Selectively Potentiates the Pathogenicity of the IgG2b and IgG3 Isotypes of a High Affinity Anti-Erythrocyte Autoantibody

Samareh Azeredo da Silveira; Shuichi Kikuchi; Liliane Fossati-Jimack; Thomas Moll; Takashi Saito; J. Sjef Verbeek; Marina Botto; Mark Walport; Michael C. Carroll; Shozo Izui

By generating four IgG isotype-switch variants of the high affinity 34–3C anti-erythrocyte autoantibody, and comparing them to the IgG variants of the low affinity 4C8 anti-erythrocyte autoantibody that we have previously studied, we evaluated in this study how high affinity binding to erythrocytes influences the pathogenicity of each IgG isotype in relation to the respective contributions of Fcγ receptor (FcγR) and complement. The 34–3C autoantibody opsonizing extensively circulating erythrocytes efficiently activated complement in vivo (IgG2a = IgG2b > IgG3), except for the IgG1 isotype, while the 4C8 IgG autoantibody failed to activate complement. The pathogenicity of the 34–3C autoantibody of IgG2b and IgG3 isotypes was dramatically higher (>200-fold) than that of the corresponding isotypes of the 4C8 antibody. This enhanced activity was highly (IgG2b) or totally (IgG3) dependent on complement. In contrast, erythrocyte-binding affinities only played a minor role in in vivo hemolytic activities of the IgG1 and IgG2a isotypes of 34–3C and 4C8 antibodies, where complement was not or only partially involved, respectively. The remarkably different capacities of four different IgG isotypes of low and high affinity anti-erythrocyte autoantibodies to activate FcγR-bearing effector cells and complement in vivo demonstrate the role of autoantibody affinity maturation and of IgG isotype switching in autoantibody-mediated pathology.


Nature Medicine | 2002

Immunoglobulin-free light chains elicit immediate hypersensitivity-like responses

Frank A. Redegeld; Maurice W. van der Heijden; M. Kool; Bianca Heijdra; Johan Garssen; Aletta D. Kraneveld; Henk van Loveren; Paul Roholl; Takashi Saito; J. Sjef Verbeek; Jill W. C. Claassens; Andries S. Koster; Frans P. Nijkamp

Immunoglobulin (Ig)-free light chains IgLC are present in serum and their production is augmented under pathological conditions such as multiple sclerosis, rheumatoid arthritis and neurological disorders. Until now, no (patho)physiological function has been ascribed to circulating Ig light chains. Here we show that IgLCs can confer mast cell–dependent hypersensitivity in mice. Antigenic stimulation results in plasma extravasation, cutaneous swelling and mast-cell degranulation. We show that IgLCs have a crucial role in development of contact sensitivity, which could be completely prevented by a novel IgLC antagonist. Although IgE and IgG1 are central to the induction of immediate hypersensitivity reactions, our results show that IgLCs have similar activity. IgLCs may therefore be a novel factor in the humoral immune response to antigen exposure. Our findings open new avenues in investigating the pathogenesis of autoimmune diseases and their treatments.


Journal of Immunology | 2011

Interaction with FcγRIIB Is Critical for the Agonistic Activity of Anti-CD40 Monoclonal Antibody

Ann L. White; H. T. Claude Chan; Ali Roghanian; Ruth R. French; C. Ian Mockridge; Alison L. Tutt; Sandra V. Dixon; Daniel Ajona; J. Sjef Verbeek; Aymen Al-Shamkhani; Mark S. Cragg; Stephen A. Beers; Martin J. Glennie

A high activatory/inhibitory FcγR binding ratio is critical for the activity of mAb such as rituximab and alemtuzumab that attack cancer cells directly and eliminate them by recruiting immune effectors. Optimal FcγR binding profiles of other anti-cancer mAb, such as immunostimulatory mAb that stimulate or block immune receptors, are less clear. In this study, we analyzed the importance of isotype and FcγR interactions in controlling the agonistic activity of the anti-mouse CD40 mAb 3/23. Mouse IgG1 (m1) and IgG2a (m2a) variants of the parental 3/23 (rat IgG2a) were engineered and used to promote humoral and cellular responses against OVA. The mouse IgG1 3/23 was highly agonistic and outperformed the parental Ab when promoting Ab (10–100-fold) and T cell (OTI and OTII) responses (2- to >10-fold). In contrast, m2a was almost completely inactive. Studies in FcγR knockout mice demonstrated a critical role for the inhibitory FcγRIIB in 3/23 activity, whereas activatory FcγR (FcγRI, -III, and -IV) was dispensable. In vitro experiments established that the stimulatory effect of FcγRIIB was mediated through Ab cross-linking delivered in trans between neighboring cells and did not require intracellular signaling. Intriguingly, activatory FcγR provided effective cross-linking of 3/23 m2a in vitro, suggesting the critical role of FcγRIIB in vivo reflects its cellular distribution and bioavailability as much as its affinity for a particular Ab isotype. In conclusion, we demonstrate an essential cross-linking role for the inhibitory FcγRIIB in anti-CD40 immunostimulatory activity and suggest that isotype will be an important issue when optimizing reagents for clinical use.


Journal of Immunology | 2000

A Codominant Role of FcγRI/III and C5aR in the Reverse Arthus Reaction

Ulrich Baumann; Jörg Köhl; Thomas Tschernig; Kirsten Schwerter-Strumpf; J. Sjef Verbeek; Reinhold E. Schmidt; J. Engelbert Gessner

Recent attempts to specify the relative contribution of FcR and complement in various experimental systems of immune complex disease have led to opposing conclusions. As concluded in IgG FcRγ−/− mice, manifestation of disease is almost exclusively determined by FcγR on effector cells, arguing for a minor role of complement. In contrast, data obtained with C5aR−/− mice suggested that, dependent on the tissue site, complement is more important than FcγR. In this paper, we demonstrate that, in response to IgG immune complex formation, FcγRI/III- and C5aR-mediated pathways are both necessary and only together are they sufficient to trigger the full expression of inflammation in skin and lung. Moreover, both effector systems are not entirely independent, suggesting an interaction between FcγR and C5aR. Therefore, FcγR-mediated responses can be integrated through C5aR activation, which may explain why these two receptor pathways have previously been considered to dominate each other.

Collaboration


Dive into the J. Sjef Verbeek's collaboration.

Top Co-Authors

Avatar

Ferry Ossendorp

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jill W. C. Claassens

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marcel Camps

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter Boross

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cor Breukel

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jos van der Kaa

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andreea Ioan-Facsinay

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge