Network


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

Hotspot


Dive into the research topics where Wendy Jansen is active.

Publication


Featured researches published by Wendy Jansen.


Journal of Experimental Medicine | 2003

A conduit system distributes chemokines and small blood-borne molecules through the splenic white pulp.

Martijn A. Nolte; Jeroen A.M. Beliën; Inge L. Schadee-Eestermans; Wendy Jansen; Wendy W. J. Unger; Nico van Rooijen; Georg Kraal; Reina E. Mebius

Access to the splenic white pulp is restricted to lymphocytes and dendritic cells. Here we show that movement of molecules from the blood into these confined areas is also limited. Large molecules, such as bovine serum albumin (68 kD), immunoglobulin G (150 kD), and 500 kD dextran are unable to enter the white pulp, whereas smaller blood-borne molecules can directly permeate this compartment. The distribution is restricted to a stromal network that we refer to as the splenic conduit system. The small lumen of the conduit contains collagen fibers and is surrounded in the T cell areas by reticular fibroblasts that express ER-TR7. It also contains the chemokine CCL21. Conversely, in B cell follicles the B cell–attracting chemokine CXCL13 was found to be associated with the conduit and absence of ER-TR7+ fibroblasts. These results show heterogeneity of reticular fibroblasts that enfold the conduit system and suggest that locally produced chemokines are transported through and presented on this reticular network. Therefore, the conduit plays a role in distribution of both blood-borne and locally produced molecules and provides a framework for directing lymphocyte migration and organization of the splenic white pulp.


Journal of Immunology | 2004

Presumptive Lymph Node Organizers are Differentially Represented in Developing Mesenteric and Peripheral Nodes

Mark F. R. Vondenhoff; Edwin J. Heeregrave; Anna E. de Weerd; Wendy Jansen; David G. Jackson; Georg Kraal; Reina E. Mebius

During murine embryogenesis, the formation of Peyer’s patches (PPs) is initiated by CD45+CD4+CD3− lymphoid tissue inducers that trigger adhesion molecule expression and specific chemokine production from an organizing stromal cell population through ligation of the lymphotoxin-β receptor. However, the steps involved in the development of lymph nodes (LNs) are less clear than those of PPs, and the characteristics of the organizing cells within the LN anlagen have yet to be documented. In this study, we show for the first time that the early anlage is bordered by an endothelial layer that retains a mixed lymphatic and blood vascular phenotype up to embryonic day 16.5. This in turn encompasses CD45+CD4+CD3− cells interspersed with ICAM-1/VCAM-1/mucosal addressin cell adhesion molecule-1, lymphotoxin-β receptor-positive, chemokine-producing cells analogous to the organizing population previously observed in PPs. Moreover, these LN organizers also express the TNF family member, TRANCE. Lastly, we show that the ICAM-1/VCAM-1/mucosal addressin cell adhesion molecule-1 cells present in peripheral and mesenteric LN form two discrete populations expressing either intermediate or high levels of these adhesion molecules but that the former population is specifically reduced in PLN. These findings provide a possible explanation for the well-known differences in developmental requirements for nodes at peripheral or mesenteric locations.


Journal of Immunology | 2009

Lymph Node Stromal Cells Support Dendritic Cell-Induced Gut-Homing of T Cells

Rosalie Molenaar; Mascha Greuter; Arnold P. J. van der Marel; Ramon Roozendaal; Stefan F. Martin; Fanny Edele; Jochen Huehn; Reinhold Förster; Tom O'Toole; Wendy Jansen; Inge L. Eestermans; Georg Kraal; Reina E. Mebius

T cells are imprinted to express tissue-specific homing receptors upon activation in tissue-draining lymph nodes, resulting in their migration to the site of Ag entry. Expression of gut-homing molecules α4β7 and CCR9 is induced by retinoic acid, a vitamin A metabolite produced by retinal dehydrogenases, which are specifically expressed in dendritic cells as well as stromal cells in mucosa-draining lymph nodes. In this study, we demonstrate that mesenteric lymph node stromal cell-derived retinoic acid can directly induce the expression of gut-homing molecules on proliferating T cells, a process strongly enhanced by bone marrow-derived dendritic cells in vitro. Therefore, cooperation of sessile lymph node stromal cells with mobile dendritic cells warrants the imprinting of tissue specific homing receptors on activated T cells.


Journal of Immunology | 2005

FcγRIIB Regulates Nasal and Oral Tolerance: A Role for Dendritic Cells

Janneke N. Samsom; Lisette A. van Berkel; Joop M. L. M. van Helvoort; Wendy W. J. Unger; Wendy Jansen; T. Thepen; Reina E. Mebius; Sjef Verbeek; Georg Kraal

Mucosal tolerance prevents the body from eliciting productive immune responses against harmless Ags that enter the body via the mucosae, and is mediated by the induction of regulatory T cells that differentiate in the mucosa-draining lymph nodes (LN) under defined conditions of Ag presentation. In this study, we show that mice deficient in FcγRIIB failed to develop mucosal tolerance to OVA, and demonstrate in vitro and in vivo a critical role for this receptor in modulating the Ag-presenting capacity of dendritic cells (DC). In vitro it was shown that absence of FcγRIIB under tolerogenic conditions led to increased IgG-induced release of inflammatory cytokines such as MCP-1, TNF-α, and IL-6 by bone marrow-derived DC, and increased their expression of costimulatory molecules, resulting in an altered immunogenic T cell response associated with increased IL-2 and IFN-γ secretion. In vivo we could show enhanced LN-DC activation and increased numbers of Ag-specific IFN-γ-producing T cells when FcγRIIB−/− mice were treated with OVA via the nasal mucosa, inferring that DC modulation by FcγRIIB directed the phenotype of the T cell response. Adoptive transfer of CD4+ T cells from the spleen of FcγRIIB−/− mice to naive acceptor mice demonstrated that OVA-responding T cells failed to differentiate into regulatory T cells, explaining the lack of tolerance in these mice. Our findings demonstrate that signaling via FcγRIIB on DC, initiated by local IgG in the mucosa-draining LN, down-regulates DC activation induced by nasally applied Ag, resulting in those defined conditions of Ag presentation that lead to Tr induction and tolerance.


Journal of Immunology | 2003

Early Events in Peripheral Regulatory T Cell Induction via the Nasal Mucosa

Wendy W. J. Unger; Femke Hauet-Broere; Wendy Jansen; Lisette A. van Berkel; Georg Kraal; Janneke N. Samsom

Nasal application of soluble Ags leads to Ag-specific suppression of systemic immune responses. This tolerance can be transferred to naive mice by CD4+ regulatory T cells (TR cells) from the spleen, but little is known about the induction of mucosal TR cells in vivo. To investigate the induction of TR cells in the nose-draining cervical lymph node (CLN), CD4+ T cells from DO11.10 OVA TCR transgenic mice were transferred to BALB/c recipients. Within 48 h after nasal OVA application, CD4+ DO11.10 T cells in CLN, but not in the peripheral lymph node, had divided. Similarly, nonmucosal (i.m.) OVA application also induced CD4+ DO11.10 T cells to proliferate in the draining inguinal lymph node (ILN), yet more vigorously and with different kinetics than the CD4+ DO11.10 T cells in CLN. Functional analysis revealed that only proliferating CD4+ DO11.10 T cells from CLN, and not ILN, could transfer tolerance to naive recipients. CD4+ DO11.10 T cells from CLN were phenotypically similar to CD4+ DO11.10 T cells from ILN, however, in CLN a higher percentage of CD25+ proliferating CD4+ DO11.10 T cells were detected compared with ILN. CD25 is not a discriminative marker for mucosal TR cells because both CD25+ and CD25− CD4+ DO11.10 T cells from the CLN could suppress delayed type hypersensitivity responses in adoptive transfer. These findings demonstrate that although striking similarities exist between the differentiation of TR and effector T cells, this does not include their function. We are the first to demonstrate that functional TR cells, which reside within both CD25+ and CD25− subsets, can be isolated from CLN as early as 3 days after nasal OVA application.


Journal of Immunology | 2004

Initiation of Cellular Organization in Lymph Nodes Is Regulated by Non-B Cell-Derived Signals and Is Not Dependent on CXC Chemokine Ligand 13

Frances E. Lund; Vu N. Ngo; Troy D. Randall; Wendy Jansen; Mascha Greuter; Rene de Waal-Malefyt; Georg Kraal; Jason G. Cyster; Reina E. Mebius

The molecular and cellular events that initiate the formation of T and B cell areas in developing lymph nodes are poorly understood. In this study we show that formation of the lymphoid architecture in murine neonatal lymph nodes evolves through a series of distinct stages. The initial segregation of T and B cells is regulated in a CXCL13-independent manner, characterized by the localization of B cells in a ring-like pattern in the outer cortex on day 4. However, during this CXCL13-independent phase of lymph node modeling, CXCL13 is expressed and regulated in a lymphotoxin-α1β2 (LTα1β2)-dependent manner. Surprisingly, neonatal B cells are unable to respond to this chemokine and also lack surface LTα1β2 expression. At this time, CD45+CD4+CD3− cells are the predominant LTα1β2-expressing cells and are also capable of responding to CXCL13. From day 4 on, architectural changes become CXCL13 dependent, and B cells become fully CXCL13 responsive, express LTα1β2, and cluster in anatomically distinct follicles. Because the initial induction of CXCL13 is dependent on LTα1β2, a role for CD45+CD4+CD3− cells in inducing chemokine expression in the developing lymph nodes is proposed and, as such, a role in initiation of the shaping of the microenvironment.


Journal of Immunology | 2007

Secretory Leukoprotease Inhibitor in Mucosal Lymph Node Dendritic Cells Regulates the Threshold for Mucosal Tolerance

Janneke N. Samsom; Arnold P. J. van der Marel; Lisette A. van Berkel; Joop M.L.M. van Helvoort; Y. Simons-Oosterhuis; Wendy Jansen; Mascha Greuter; Rob L. H. Nelissen; Cees M. L. Meeuwisse; Edward E. S. Nieuwenhuis; Reina E. Mebius; Georg Kraal

The notion that the mucosal immune system maintains a tolerogenic response to harmless Ags while continually being challenged with microbial products seems an enigma. The aim of this study was to unravel mechanisms that are involved in regulating the development of tolerance under constant microbial pressure. The tolerogenic response to Ags administered via the nasal mucosa is dependent on the organized lymphoid tissue of the cervical lymph nodes (LN). We show that cervical LN differentially express secretory leukoprotease inhibitor (SLPI) compared with peripheral LN. SLPI was expressed by dendritic cells (DCs) and because SLPI is known to suppress LPS responsiveness, it was hypothesized that its expression in mucosal DCs may be required to regulate cellular activation to microbial products. Indeed, compared with wild-type controls, bone marrow-derived DCs from SLPI−/− mice released more inflammatory cytokines and enhanced T cell proliferation after stimulation with low dose LPS. This increased sensitivity to LPS was accompanied by increased NF-κB p65 activation in SLPI−/− DCs. In vivo, nasal application of OVA with LPS to SLPI−/− mice resulted in enhanced DC activation in the cervical LN reflected by increased costimulatory molecule expression and release of inflammatory cytokines. This led to failure to maintain tolerance to nasal OVA application in the presence of low doses of LPS. We propose that expression of SLPI functions as a rheostat by controlling the level of bacterial stimuli that induce mucosal DC activation. As such, it regulates the quality of the ensuing Ag-specific immune response in the mucosa draining LN.


Allergy | 2004

Preferential expression of IgG2b in nose draining cervical lymph nodes and its putative role in mucosal tolerance induction

J. M. L. M. Van Helvoort; Janneke N. Samsom; D. Chantry; Wendy Jansen; I. Schadee‐Eestermans; T. Thepen; Reina E. Mebius; Georg Kraal

Induction of intranasal tolerance prevents the body from eliciting unwanted immune responses against harmless antigens that enter the body through the nasal mucosa. To study the intrinsic capacities of the cervical, nose draining lymph nodes (CLN), which are essential for tolerance induction, genes that are differentially expressed in CLN and not in peripheral lymph nodes (PLN) were characterized. The gene that is predominantly overexpressed in CLN codes for IgG2b. This is confirmed by a higher percentage of IgG2b+ B220+ cells in CLN compared with any PLN. However, this predominance of IgG2b‐positive B cells in the CLN is not specific for the lymph node itself but rather determined by the region drained by lymph nodes at the cervical site, as transplanted PLN at these locations show a comparable predominance. It was demonstrated that IgG2b, when compared with IgG1, led to differential activation of dendritic cells (DC) through Fc receptor signalling. The results point to a unique local combination of cells and factors in the nose draining CLN leading to highly specialized immune reactivity. The results point out that predominance of a distinct IgG isotype in a lymphoid environment may lead to highly specialized immune reactivity.


Immunity | 2004

Induction of Secondary and Tertiary Lymphoid Structures in the Skin

Wendy Jansen; Georg Kraal; Reina E. Mebius


International Immunology | 2003

Nasal tolerance induces antigen-specific CD4^+CD25^- regulatory T cells that can transfer their regulatory capacity to naive CD4^+ T cells

Wendy W. J. Unger; Wendy Jansen; Danielle A. W. Wolvers; Astrid G. S. van Halteren; Georg Kraal; Janneke N. Samsom

Collaboration


Dive into the Wendy Jansen's collaboration.

Top Co-Authors

Avatar

Georg Kraal

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Reina E. Mebius

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Janneke N. Samsom

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Wendy W. J. Unger

VU University Medical Center

View shared research outputs
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