Network


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

Hotspot


Dive into the research topics where Anne Christine W. Vos is active.

Publication


Featured researches published by Anne Christine W. Vos.


Stem Cells | 2011

Pretreatment with interferon-γ enhances the therapeutic activity of mesenchymal stromal cells in animal models of colitis

Marjolijn Duijvestein; Manon E. Wildenberg; Mick M. Welling; Simone D. Hennink; Ilse Molendijk; Vanessa‑Leigh van Zuylen; Tjalling Bosse; Anne Christine W. Vos; Eveline S.M. de Jonge-Muller; Helene Roelofs; Louise van der Weerd; Hein W. Verspaget; Willem E. Fibbe; Anje A. te Velde; Gijs R. van den Brink; Daniel W. Hommes

Mesenchymal stromal cells (MSCs) are currently under investigation for the treatment of inflammatory disorders, including Crohns disease. MSCs are pluripotent cells with immunosuppressive properties. Recent data suggest that resting MSCs do not have significant immunomodulatory activity, but that the immunosuppressive function of MSCs has to be elicited by interferon‐γ (IFN‐γ). In this article, we assessed the effects of IFN‐γ prestimulation of MSCs (IMSCs) on their immunosuppressive properties in vitro and in vivo. To this end, we pretreated MSCs with IFN‐γ and assessed their therapeutic effects in dextran sodium sulfate (DSS)‐ and trinitrobenzene sulfonate (TNBS)‐induced colitis in mice. We found that mice treated with IMSCs (but not MSCs) showed a significantly attenuated development of DSS‐induced colitis. Furthermore, IMSCs alleviated symptoms of TNBS‐induced colitis. IMSC‐treated mice displayed an increase in body weight, lower colitis scores, and better survival rates compared with untreated mice. In addition, serum amyloid A protein levels and local proinflammatory cytokine levels in colonic tissues were significantly suppressed after administration of IMSC. We also observed that IMSCs showed greater migration potential than unstimulated MSCs to sites within the inflamed intestine. In conclusion, we show that prestimulation of MSCs with IFN‐γ enhances their capacity to inhibit Th1 inflammatory responses, resulting in diminished mucosal damage in experimental colitis. These data demonstrate that IFN‐γ activation of MSCs increases their immunosuppresive capacities and importantly, their therapeutic efficacy in vivo. STEM CELLS 2011;29:1549–1558


Inflammatory Bowel Diseases | 2012

Regulatory macrophages induced by infliximab are involved in healing in vivo and in vitro.

Anne Christine W. Vos; Manon E. Wildenberg; Ingrid Arijs; Marjolijn Duijvestein; Auke Verhaar; Gert De Hertogh; Severine Vermeire; Paul Rutgeerts; Gijs R. van den Brink; Daniel W. Hommes

Background: Regulatory macrophages play an important role in wound healing and gut homeostasis and have antiinflammatory properties. Induction of this cell type (M&psgr;ind) by the anti‐tumor necrosis factor (TNF) antibodies, infliximab and adalimumab, has recently been shown in vitro. Also, the superiority of infliximab/azathioprine combination therapy over infliximab or azathioprine monotherapy has recently been established, but the mechanism behind this remains unclear. The aim of this study was to examine the induction of regulatory macrophages in patients with and without mucosal healing in response to infliximab. In addition, we studied the effect of infliximab/azathioprine combination treatment on the differentiation and function of regulatory macrophages. Methods: Inflammatory bowel disease (IBD) patients (n = 10) underwent endoscopy before and after first infliximab treatment. Immunohistochemical staining of CD68 and CD206 was performed in all patients. Mixed lymphocyte reactions (MLRs) were treated with infliximab, azathioprine, or both. Macrophage phenotype was evaluated by flow cytometry and inhibition of T‐cell proliferation was measured in a secondary MLR containing macrophages and third‐party lymphocytes. Results: A significant induction of regulatory macrophages was observed in patients with mucosal healing after treatment with infliximab; this induction was absent in patients without mucosal healing. In addition, M&psgr;ind have the ability to induce wound healing in an in vitro model, further suggesting a key role for infliximab‐induced macrophages in mucosal healing. Upon infliximab/azathioprine combination treatment, an increased number of regulatory macrophages was observed. These macrophages also displayed stronger immunosuppressive properties than macrophages induced by infliximab monotherapy. Conclusions: These data show that regulatory macrophages may be involved in mucosal healing and provide a rationale for the superiority of infliximab/azathioprine combination treatment observed in the clinic. (Inflamm Bowel Dis 2012;)


Inflammatory Bowel Diseases | 2011

Risk of Malignant Lymphoma in Patients with Inflammatory Bowel Diseases: A Dutch Nationwide Study

Anne Christine W. Vos; N. Bakkal; Robert C. Minnee; M.K. Casparie; D.J. de Jong; Gerard Dijkstra; Pieter Stokkers; A.A. van Bodegraven; Marieke Pierik; C.J. van der Woude; Bas Oldenburg; Daan W. Hommes

Background: Immune suppressant medications such as thiopurines and anti‐tumor necrosis factor agents are important for maintaining disease control in most patients with inflammatory bowel diseases (IBDs); however, their use has been associated with the development of malignant lymphoma. The purpose of this Dutch nationwide study was to estimate the relative risk of malignant lymphoma in IBD patients. Methods: IBD patients who developed a lymphoma between 1997 and 2004 were identified using the Dutch National Database of PALGA. Data from confirmed cases were collected from individual hospitals, including data on Epstein–Barr virus (EBV). The age‐adjusted 8‐year incidence of malignant lymphoma in the Netherlands was retrieved from the Central Bureau of Statistics. Results: Forty‐two hospitals were visited and 285 matches evaluated in the total cohort of 17,834 IBD patients. Forty‐four lymphomas were observed, resulting in a relative risk of 1.27 (95% confidence interval [CI]: 0.92–1.68). Only 19 of 44 patients (43%) were exposed to azathioprine/6‐mercaptopurine (AZA/6‐MP). Remarkably, 92% of patients (11/12) with EBV‐positive lymphoma used AZA/6‐MP, in contrast to only 19% patients (4/21) with EBV‐negative lymphoma, suggesting a strong relation between EBV‐positive lymphoma and thiopurine use. Conclusions: This nationwide study does not suggest a significant overall increased risk for lymphoma in IBD patients. A distinct correlation between EBV‐positive lymphoma and AZA/6‐MP use was observed. (Inflamm Bowel Dis 2011;)


Gastroenterology | 2012

Autophagy Attenuates the Adaptive Immune Response by Destabilizing the Immunologic Synapse

Manon E. Wildenberg; Anne Christine W. Vos; Simone C. Wolfkamp; Marjolijn Duijvestein; Auke Verhaar; Anje A. te Velde; Gijs R. van den Brink; Daniel W. Hommes

BACKGROUND & AIMS Variants in the genes ATG16L1 and IRGM affect autophagy and are associated with the development of Crohns disease. It is not clear how autophagy is linked to loss of immune tolerance in the intestine. We investigated the involvement of the immunologic synapse-the site of contact between dendritic cells (DCs) and T cells, which contains molecules involved in antigen recognition and regulates immune response. METHODS DC autophagy was reduced using small interfering RNAs or pharmacologic inhibitors. DC phenotype and function were analyzed by confocal microscopy, time-lapse microscopy, and flow cytometry. We also examined DCs isolated from patients with Crohns disease who carried the ATG16L1 risk allele. RESULTS Immunologic synapse formation induced formation of autophagosomes in DCs; the autophagosomes were oriented toward the immunologic synapse and contained synaptic components. Knockdown of ATG16L1 and IRGM with small interfering RNAs in DCs resulted in hyperstable interactions between DCs and T cells, increased activation of T cells, and activation of a T-helper 17 cell response. LKB1 was recruited to the immunologic synapse, and induction of autophagy in DC required inhibition of mammalian target of rapamycine signaling by the LKB1-AMP activated protein kinase (AMPK) pathway. DCs from patients with Crohns disease who had an ATG16L1 risk allele had a similar hyperstability of the immunologic synapse. CONCLUSIONS Autophagy is induced upon formation of the immunologic synapse and negatively regulates T-cell activation. This mechanism might increase adaptive immunity in patients with Crohns disease who carry ATG16L1 risk alleles.


Journal of Crohns & Colitis | 2013

Impaired autophagy leads to abnormal dendritic cell–epithelial cell interactions

Caterina Strisciuglio; Marjolijn Duijvestein; Auke Verhaar; Anne Christine W. Vos; Gijs R. van den Brink; Daniel W. Hommes; Manon E. Wildenberg

BACKGROUND AND AIMS Dendritic cells (DC) are key players in intestinal immunity, as these cells can direct the immune response to either a tolerogenic or an immunogenic phenotype. In the intestine, DC sample and process luminal antigens by protruding dendrites through the epithelial cell layer. At the same time barrier integrity is maintained through the continuous formation of tight junctions. Aberrations in these interactions may lead to altered antigen sampling and improper immune responses. We have recently shown that autophagy, a process implicated in the pathogenesis of Crohns disease, regulates cellular interactions in the context of DC and T cells. In this study we aimed to determine whether autophagy also regulates DC-epithelial cell interactions and whether this influences the ensuing immune response. METHODS DC were generated from peripheral blood monocytes of healthy volunteers. For interaction studies, DC were co-cultured with intestinal epithelial cells on the baso-lateral side of a transwell insert. Modulation of autophagy was achieved using atg16l1 specific siRNA or pharmacological inhibitors. Intraepithelial protrusion of dendrites was determined by confocal microscopy. Luminal sampling and DC activation status were analyzed by flow cytometry. Protein expression was measured by immunoblotting and cytometric bead assay. RESULTS Adhesion molecules E-cadherin and occludin partly localized to autophagosomes and increased autophagy resulted in decreased levels of these proteins. Reduced autophagy in either DC, epithelial cells or both resulted in the decreased formation of transepithelial protrusions by DC as well as a reduction in antigen sampling. Moreover, when autophagy was inhibited in the co-culture model, DC expressed increased levels of HLA-DR and costimulatory molecule CD86. Furthermore, decreased levels of autophagy resulted in lower IL-10 production by DC and these cells induced significantly more T-cell proliferation in an allogeneic mixed lymphocyte reaction. CONCLUSIONS In intestinal DC-epithelial cell interactions, autophagy deficiency leads to decreased antigen sampling, increased DC maturation and a more pro-inflammatory type of DC.


Cytotherapy | 2011

Mesenchymal stromal cell function is not affected by drugs used in the treatment of inflammatory bowel disease

Marjolijn Duijvestein; Ilse Molendijk; Helene Roelofs; Anne Christine W. Vos; Auke Verhaar; Marlies E.J. Reinders; Willem E. Fibbe; Hein W. Verspaget; Gijs R. van den Brink; Manon E. Wildenberg; Daniel W. Hommes

BACKGROUND AND AIMS Mesenchymal stromal cells (MSC) have both multilineage differentiation capacity and immunosuppressive properties. Promising results with MSC administration have been obtained in experimental colitis. Clinical application of MSC for the treatment of inflammatory bowel disease (IBD) is currently under investigation in phase I-III trials in patients with past or concurrent immunomodulating therapy. However, little is known about MSC interactions with these immunosuppressive drugs. To address this issue we studied the combined effect of MSC and IBD drugs in in vitro functionality assays. METHODS The effects of azathioprine, methotrexate, 6-mercaptopurine and anti-tumor necrosis factor (TNF)-α on MSC phenotype, survival, differentiation capacity and immunosuppressive capacity were studied. RESULTS MSC exposed to physiologically relevant concentrations of IBD drugs displayed a normal morphology and fulfilled phenotypic and functional criteria for MSC. Differentiation into adipocyte and osteocyte lineages was not affected and cells exhibited normal survival after exposure to the various drugs. MSC suppression of peripheral blood mononuclear cell (PBMC) proliferation in vitro was not hampered by IBD drugs. In fact, in the presence of 6-mercaptopurine and anti-TNF-α antibodies, the inhibitory effect of this drug alone was enhanced, suggesting an additive effect of pharmacotherapy and MSC treatment. CONCLUSIONS This study demonstrates that, in vitro, MSC phenotype and function are not affected by therapeutic concentrations of drugs commonly used in the treatment of IBD. These findings are important for the potential clinical use of MSC in combination with immunomodulating drugs and anti-TNF-α therapy.


Journal of Crohns & Colitis | 2016

Autophagy Contributes to the Induction of Anti-TNF Induced Macrophages

Alon D. Levin; Pim J. Koelink; Felicia M. Bloemendaal; Anne Christine W. Vos; Geert D’Haens; Gijs R. van den Brink; Manon E. Wildenberg

BACKGROUND AND AIMS Anti-tumour necrosis factor [TNF] antibodies induce regulatory macrophages which display a phenotype resembling M2 type macrophages. Anti-TNF induced macrophages [Mϕind] have immunosuppressive and wound healing properties. The factors that contribute to the induction of Mϕind remain to be explored. Autophagy has been described as a factor that is important for the induction and function of M2 type macrophages. We studied the contribution of autophagy to the induction of Mϕind. METHODS We studied the effect of autophagy on Mϕind in vitro using peripheral blood mononuclear cells. Interferon gamma [IFN-γ] induced macrophages [Mφ1] were generated by culturing monocytes in the presence of IFN-γ. Mϕind were generated by performing mixed lymphocyte reactions [MLR] in the presence of anti-TNF antibodies; 28 healthy donors were genotyped for rs_2241880 [ATG16L1]. Cells were analysed by autophagy gene array, immunofluorescence, western blot, flowcytometry, 3H-thymidine incorporation and MTS assay. RESULTS Mϕind had a different expression profile of autophagy related transcripts with increased expression of 33/40 altered genes compared with Mφ1. In addition, autophagic activity was increased in Mϕind compared with Mφ1. Induction of Mϕind was positively correlated to the number of wild-type alleles for the ATG16L1 T300A risk allele present in the culture. Finally, the autophagy-related protein cathepsin S was highly expressed in Mφind and inhibition resulted in decreased viability as well as decreased expression of CD206. CONCLUSIONS Mϕind have increased levels of autophagy compared with inflammatory Mφ1, and the induction of these macrophages is impaired in donors carrying the T300A risk allele for the ATG16L1. Given the association between Mϕind and clinical response, this suggests that an intact autophagy pathway may be important for an optimal response to anti-TNF therapy in inflammatory bowel disease.


Inflammatory Bowel Diseases | 2013

Miltefosine suppresses inflammation in a mouse model of inflammatory bowel disease

Auke Verhaar; Manon E. Wildenberg; Anje A. te Velde; Sybren L. Meijer; Anne Christine W. Vos; Marjolijn Duijvestein; Maikel P. Peppelenbosch; Daniel W. Hommes; Gijs R. van den Brink

Background:The repertoire of immunomodulators that can be used for the treatment of inflammatory bowel disease is limited. The use of these drugs is further restricted by the occurrence of side effects in a proportion of patients. Miltefosine (hexadecylphosphocholine) is a lipid drug developed in the 1980s for the treatment of cancer but is nowadays best known for its application in the oral treatment of leishmaniasis. Although the exact mechanism of action of miltefosine has yet to be elucidated, the drug has previously been shown to inhibit phospholipases and protein kinase C, both key components of proproliferative signal transduction in T cells. Methods:Stimulated peripheral blood lymphocyte were treated with miltefosine, and proliferation was measured. We use the CD45RBhigh T-cell transfer colitis model to investigate the effect of miltefosine treatment on intestinal inflammation. Effects on the severity of colitis were studied by histochemical and immunohistochemical staining, and cytokine levels were determined using a cytokine bead array. Results:Miltefosine inhibited T-cell proliferation in vitro. In the transfer model, miltefosine significantly ameliorated the severity of colitis as measured by clinical, (immuno)histochemical, and biochemical parameters. Conclusions:Miltefosine inhibits T-cell proliferation and effectively reduces inflammation in the T-cell transfer model. The drug may therefore be a candidate immunomodulator for inflammatory bowel disease.


Journal of Immunology | 2013

Superantigen-Induced Steroid Resistance Depends on Activation of Phospholipase Cβ2

Auke Verhaar; Manon E. Wildenberg; Marjolijn Duijvestein; Anne Christine W. Vos; Maikel P. Peppelenbosch; Mark Löwenberg; Daniel W. Hommes; Gijs R. van den Brink

The glucocorticoid receptor is present in a TCR-associated complex, which includes the Src family tyrosine kinase Lck. Glucocorticoids rapidly dissociate this complex, resulting in the inhibition of canonical Lck-phospholipase C (PLC)γ–dependent TCR signaling. The relative importance of this nongenomic role for the glucocorticoid receptor compared with its direct transcriptional effects is not known. Superantigens induce a state of steroid resistance in activated T cells. It was reported that, in addition to canonical Lck-PLCγ signaling, superantigens can activate a noncanonical G protein–PLCβ–dependent signaling pathway. In this study, we show that staphylococcal enterotoxin B activates a Gαq and PLCβ2–dependent pathway in human T cells. We find that this pathway bypasses the need for canonical Lck-PLCγ signaling in T cell activation and renders superantigen-stimulated T cells insensitive to glucocorticoids in vitro. We show that the PLCβ inhibitor U-73122 sensitizes staphylococcal enterotoxin B–treated mice to dexamethasone in vivo. In conclusion, we find that effects of glucocorticoids on TCR-induced T cell proliferation are mainly nongenomic and can be bypassed by the activation of an Lck-independent signaling pathway.


Archive | 2012

Step-Up vs. Top-Down Approach in Medical Management of Inflammatory Bowel Disease

Anne Christine W. Vos; Daniel W. Hommes

During recent years, the treatment for patients with Crohn’s disease (CD) has changed substantially. With the introduction of anti-TNF alpha antibodies, the treatment goals have been more ambitious, for instance, with the introduction of mucosal healing. Today, upon diagnosing CD, clinicians are able to use predictors of a complicated disease course and act accordingly when designing an individual medical strategy.

Collaboration


Dive into the Anne Christine W. Vos's collaboration.

Top Co-Authors

Avatar

Manon E. Wildenberg

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marjolijn Duijvestein

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Auke Verhaar

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daan W. Hommes

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helene Roelofs

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Willem E. Fibbe

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge