Network


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

Hotspot


Dive into the research topics where Marjolein E. Sanders is active.

Publication


Featured researches published by Marjolein E. Sanders.


Journal of Immunology | 2004

Rap1 Signaling Is Required for Suppression of Ras-Generated Reactive Oxygen Species and Protection Against Oxidative Stress in T Lymphocytes

Philip H. J. Remans; Sonja I. Gringhuis; Jacob M. van Laar; Marjolein E. Sanders; Ellen A. M. Papendrecht-van der Voort; Fried J. T. Zwartkruis; E. W. Nivine Levarht; Marcela Rosas; Paul J. Coffer; Ferdinand C. Breedveld; Johannes L. Bos; Paul P. Tak; Cornelis L. Verweij; Kris A. Reedquist

Transient production of reactive oxygen species (ROS) plays an important role in optimizing transcriptional and proliferative responses to TCR signaling in T lymphocytes. Conversely, chronic oxidative stress leads to decreased proliferative responses and enhanced transcription of inflammatory gene products, and is thought to underlie the altered pathogenic behavior of T lymphocytes in some human diseases, such as rheumatoid arthritis (RA). Although the signaling mechanisms regulating ROS production in T lymphocytes has not been identified, activation of the small GTPase Ras has been shown to couple agonist stimulation to ROS production in other cell types. We find that Ras signaling via Ral stimulates ROS production in human T lymphocytes, and is required for TCR and phorbol ester-induced ROS production. The related small GTPase Rap1 suppresses agonist, Ras and Ral–dependent ROS production through a PI3K–dependent pathway, identifying a novel mechanism by which Rap1 can distally antagonize Ras signaling pathways. In synovial fluid T lymphocytes from RA patients we observed a high rate of endogenous ROS production, correlating with constitutive Ras activation and inhibition of Rap1 activation. Introduction of dominant-negative Ras into synovial fluid T cells restored redox balance, providing evidence that deregulated Ras and Rap1 signaling underlies oxidative stress and consequent altered T cell function observed in RA.


Analytical Chemistry | 2010

TOF-Secondary Ion Mass Spectrometry Imaging of Polymeric Scaffolds with Surrounding Tissue after in Vivo Implantation

Leendert A. Klerk; Patricia Y. W. Dankers; Eliane R. Popa; Anton Willem Bosman; Marjolein E. Sanders; Kris A. Reedquist; Ron M. A. Heeren

Supramolecular polymeric materials are of increasing interest for the use as drug delivery carriers. A thorough insight in the biocompatibility and the degradation of these materials in vivo are of fundamental importance to further their development and application in medical practice. Molecular imaging techniques are powerful tools that enable the elucidation of molecular distributions in and around such polymer implants. A supramolecular polymeric hydrogel was implanted under the renal capsule to study its biocompatibility with TOF-SIMS. This results in a molecular cartography of the polymer implant combined with the cellular signature of the implantation environment. In this experiment, molecular signals are observed from cells that are involved in the biological response to the implant, e.g., macrophages. These molecular signatures are compared with macrophage standards cultured in different polarization environments. On the basis of this comparison, information can be acquired on the various macrophage differentiations that are connected to different stages in the foreign body response. Mass spectrometric imaging techniques offer the opportunity to visualize different histological phenomena in a single experiment without the need for specific immunohistochemical markers. Cellular infiltration into the polymer is visualized, offering a clear view on both biological and polymer features in a single imaging experiment.


Arthritis Research & Therapy | 2010

A Rac1 inhibitory peptide suppresses antibody production and paw swelling in the murine collagen-induced arthritis model of rheumatoid arthritis

Joana Rf Abreu; Wendy Dontje; Sarah Krausz; Daphne de Launay; Paula B. van Hennik; Anne-Marieke van Stalborch; Jean-Paul ten Klooster; Marjolein E. Sanders; Kris A. Reedquist; Margriet J. Vervoordeldonk; Peter L. Hordijk; Paul P. Tak

IntroductionThe Rho family GTPase Rac1 regulates cytoskeletal rearrangements crucial for the recruitment, extravasation and activation of leukocytes at sites of inflammation. Rac1 signaling also promotes the activation and survival of lymphocytes and osteoclasts. Therefore, we assessed the ability of a cell-permeable Rac1 carboxy-terminal inhibitory peptide to modulate disease in mice with collagen-induced arthritis (CIA).MethodsCIA was induced in DBA/1 mice, and in either early or chronic disease, mice were treated three times per week by intraperitoneal injection with control peptide or Rac1 inhibitory peptide. Effects on disease progression were assessed by measurement of paw swelling. Inflammation and joint destruction were examined by histology and radiology. Serum levels of anti-collagen type II antibodies were measured by enzyme-linked immunosorbent assay. T-cell phenotypes and activation were assessed by fluorescence-activated cell sorting analysis. Results were analyzed using Mann-Whitney U and unpaired Student t tests.ResultsTreatment of mice with Rac1 inhibitory peptide resulted in a decrease in paw swelling in early disease and to a lesser extent in more chronic arthritis. Of interest, while joint destruction was unaffected by Rac1 inhibitory peptide, anti-collagen type II antibody production was significantly diminished in treated mice, in both early and chronic arthritis. Ex vivo, Rac1 inhibitory peptide suppressed T-cell receptor/CD28-dependent production of tumor necrosis factor α, interferon γ and interleukin-17 by T cells from collagen-primed mice, and reduced induction of ICOS and CD154, T-cell costimulatory proteins important for B-cell help.ConclusionsThe data suggest that targeting of Rac1 with the Rac1 carboxy-terminal inhibitory peptide may suppress T-cell activation and autoantibody production in autoimmune disease. Whether this could translate into clinically meaningful improvement remains to be shown.


Arthritis Research & Therapy | 2009

The Ras guanine nucleotide exchange factor RasGRF1 promotes matrix metalloproteinase-3 production in rheumatoid arthritis synovial tissue

Joana Rf Abreu; Daphne de Launay; Marjolein E. Sanders; Aleksander M. Grabiec; Marleen G. H. van de Sande; Paul P. Tak; Kris A. Reedquist

IntroductionFibroblast-like synoviocytes (FLS) from rheumatoid arthritis (RA) patients share many similarities with transformed cancer cells, including spontaneous production of matrix metalloproteinases (MMPs). Altered or chronic activation of proto-oncogenic Ras family GTPases is thought to contribute to inflammation and joint destruction in RA, and abrogation of Ras family signaling is therapeutic in animal models of RA. Recently, expression and post-translational modification of Ras guanine nucleotide releasing factor 1 (RasGRF1) was found to contribute to spontaneous MMP production in melanoma cancer cells. Here, we examine the potential relationship between RasGRF1 expression and MMP production in RA, reactive arthritis, and inflammatory osteoarthritis synovial tissue and FLS.MethodsExpression of RasGRF1, MMP-1, MMP-3, and IL-6 was detected in synovial tissue by immunohistochemistry and stained sections were evaluated by digital image analysis. Expression of RasGRF1 in FLS and synovial tissue was also assessed by immunoblotting. Double staining was performed to detect proteins in specific cell populations, and cells producing MMP-1 and MMP-3. RasGRF1 expression was manipulated in RA FLS by cDNA transfection and gene silencing, and effects on MMP-1, TIMP-1, MMP-3, IL-6, and IL-8 production measured by ELISA.ResultsExpression of RasGRF1 was significantly enhanced in RA synovial tissue, and detected in FLS and synovial macrophages in situ. In cultured FLS and synovial biopsies, RasGRF1 was detected by immunoblotting as a truncated fragment lacking its negative regulatory domain. Production of MMP-1 and MMP-3 in RA but not non-RA synovial tissue positively correlated with expression of RasGRF1 and co-localized in cells expressing RasGRF1. RasGRF1 overexpression in FLS induced production of MMP-3, and RasGRF1 silencing inhibited spontaneous MMP-3 production.ConclusionsEnhanced expression and post-translational modification of RasGRF1 contributes to MMP-3 production in RA synovial tissue and the semi-transformed phenotype of RA FLS.


Journal of Immunology | 2008

Homeostatic intracellular-free Ca2+ is permissive for Rap1-mediated constitutive activation of alpha4 integrins on eosinophils.

Laurien H. Ulfman; Vera M. Kamp; Corneli W. van Aalst; Liesbeth P. Verhagen; Marjolein E. Sanders; Kris A. Reedquist; Miranda Buitenhuis; Leo Koenderman

Although much progress has been made in understanding the molecular mechanisms underlying agonist-induced “inside-out” activation of integrins, little is known about how basal levels of integrin function are maintained. This is particularly important for nonactivated eosinophils, where intermediate activation of α4β1 integrin supports recruitment to endothelial cells under flow conditions. Depletion of intracellular Ca2+ and pharmacological inhibition of phospholipase C (but not other intracellular signaling molecules, including PI3K, ERK1/2, p38 MAPK, and tyrosine kinase activity) abrogated basal α4 integrin activity in nonactivated eosinophils. Basal α4 integrin activation was associated with activation of the small GTPase Rap1, a known regulator of agonist-induced integrin function. Basal Rap activation was dependent upon phospholipase C, but not intracellular Ca2+. However, depletion of intracellular Ca2+ in CD34+ hematopoietic progenitor cells abolished RapV12-mediated induction of α4 integrin activity. Thus, residual Rap activity or constitutively active Rap activity in Ca2+-depleted cells is not sufficient to induce α4 integrin activation. These data suggest that activation of functional α4 integrin activity in resting eosinophils is mediated by Rap1 provided that the intracellular-free Ca2+ is at a normal homeostatic concentration.


American Journal of Pathology | 2010

Silencing the Expression of Ras Family GTPase Homologues Decreases Inflammation and Joint Destruction in Experimental Arthritis

Daphne de Launay; Jeroen Vreijling; Linda M. Hartkamp; Olga N. Karpus; Joana Rf Abreu; Marjolein A. van Maanen; Marjolein E. Sanders; Aleksander M. Grabiec; Jörg Hamann; Henrik Ørum; Margriet J. Vervoordeldonk; Kees Fluiter; Paul P. Tak; Kris A. Reedquist

Changes in the expression and activation status of Ras proteins are thought to contribute to the pathological phenotype of stromal fibroblast-like synoviocytes (FLS) in rheumatoid arthritis, a prototypical immune-mediated inflammatory disease. Broad inhibition of Ras and related proteins has shown protective effects in animal models of arthritis, but each of the Ras family homologues (ie, H-, K-, and N-Ras) makes distinct contributions to cellular activation. We examined the expression of each Ras protein in synovial tissue and FLS obtained from patients with rheumatoid arthritis and other forms of inflammatory arthritis. Each Ras protein was expressed in synovial tissue and cultured FLS. Each homolog was also activated following FLS stimulation with tumor necrosis factor-α or interleukin (IL)-1β. Constitutively active mutants of each Ras protein enhanced IL-1β-induced FLS matrix metalloproteinase-3 production, while only active H-Ras enhanced IL-8 production. Gene silencing demonstrated that each Ras protein contributed to IL-1β-dependent IL-6 production, while H-Ras and N-Ras supported IL-1β-dependent matrix metalloproteinase-3 and IL-8 production, respectively. The overlap in contributions of Ras homologues to FLS activation suggests that broad targeting of Ras GTPases in vivo suppresses global inflammation and joint destruction in arthritis. Consistent with this, simultaneous silencing of H-Ras, K-Ras, and N-Ras expression significantly reduces inflammation and joint destruction in murine collagen-induced arthritis, while specific targeting of N-Ras alone is less effective in providing clinical benefits.


Annals of the Rheumatic Diseases | 2010

Macrophage stimulation by angiopoietins-1 and -2 promotes recruitment and retention of white blood cells at sites of inflammation

Sarah Krausz; Marjolein E. Sanders; P P Tak; Kris A. Reedquist

The authors have previously found that macrophages are predominant targets of the angiogenic factors angiopoietin (Ang)-1 and -2 in synovial tissue of patients with rheumatoid arthritis (RA). How macrophage Tie2 ligation by Ang might contribute to disease in RA is currently …


Annals of the Rheumatic Diseases | 2010

Silencing expression of Ras family GTPase homologues decreases inflammation and joint destruction in experimental arthritis

D de Launay; Jeroen Vreijling; Joana Rf Abreu; M A van Maanen; Marjolein E. Sanders; Margriet J. Vervoordeldonk; H Oerum; Kees Fluiter; P P Tak; Kris A. Reedquist

Changes in expression and activation of Ras proteins are thought to contribute to the pathological phenotype of stromal fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA). Here we examined the expression of each Ras protein in the synovial tissue of patients with RA and disease controls and the contributions of H-, K- and N-Ras homologues to FLS activation in vitro and experimental arthritis in vivo. Synovial Ras …


Rheumatology | 2007

Blocking the receptor for C5a in patients with rheumatoid arthritis does not reduce synovial inflammation

Clarissa E. Vergunst; Danielle M. Gerlag; L. Schulz; Marjolein Vinkenoog; Tjm Smeets; Marjolein E. Sanders; Kris A. Reedquist; P P Tak


Arthritis & Rheumatism | 2007

Inhibition of forkhead box class O family member transcription factors in rheumatoid synovial tissue

J. Ludikhuize; D de Launay; D. Groot; Tom J. M. Smeets; M. Vinkenoog; Marjolein E. Sanders; Sander W. Tas; P P Tak; Kris A. Reedquist

Collaboration


Dive into the Marjolein E. Sanders's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul P. Tak

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar

P P Tak

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah Krausz

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ferdinand C. Breedveld

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge