Network


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

Hotspot


Dive into the research topics where Sarah Roord is active.

Publication


Featured researches published by Sarah Roord.


Proceedings of the National Academy of Sciences of the United States of America | 2004

Epitope-specific immunotherapy induces immune deviation of proinflammatory T cells in rheumatoid arthritis

Berent J. Prakken; Rodrigo Samodal; Tho Le; Francesca Giannoni; Gisella Puga Yung; John Scavulli; Diane Amox; Sarah Roord; Ismé de Kleer; Dustan Bonnin; Paola Lanza; Charles C. Berry; Margherita Massa; Rosario Billetta; Salvatore Albani

Modulation of epitope-specific immune responses would represent a major addition to available therapeutic options for many autoimmune diseases. The objective of this work was to induce immune deviation by mucosal peptide-specific immunotherapy in rheumatoid arthritis (RA) patients, and to dissect the related immunological mechanisms by using a technology for the detection of low-affinity class II-restricted peptide-specific T cells. A group of patients with early RA was treated for 6 months orally with dnaJP1, a peptide that induces proinflammatory T cell responses in naive RA patients. Immunological analysis at initial, intermediate and end treatment points showed an intriguing change from proinflammatory to regulatory T cell function. In fact, dnaJP1-induced T cell production of IL-4 and IL-10 increased significantly when initial and end treatment points were compared, whereas dnaJP1-induced T cell proliferation and production of IL-2, IFN-γ, and tumor necrosis factor-α decreased significantly. The total number of dnaJP1-specific cells did not change over time, whereas expression of foxP3 by CD4+CD25bright cells increased, suggesting that the treatment affected regulatory T cell function. Thus, rather than clonal deletion, the observed change in immune reactivity to dnaJP1 was the outcome of treatment-induced emergence of T cells with a different functional phenotype. This study contributes to our knowledge of mechanisms and tools needed for antigen-specific immune modulation in humans, thus laying the foundation for exploitation of this approach for therapeutic purposes.


Journal of Immunology | 2002

Immunostimulatory DNA Sequences Influence the Course of Adjuvant Arthritis

Arash Ronaghy; Berent Prakken; Kenji Takabayashi; Gary S. Firestein; David L. Boyle; Nathan J. Zvailfler; Sarah Roord; Salvatore Albani; Dennis A. Carson; Eyal Raz

Bacterial DNA is enriched in unmethylated CpG motifs that have been shown to activate the innate immune system. These immunostimulatory DNA sequences (ISS) induce inflammation when injected directly into joints. However, the role of bacterial DNA in systemic arthritis is not known. The purpose of the present experiments was to determine whether ISS contributes to the development of adjuvant arthritis in Lewis rats after intradermal injection of heat-killed Mycobacterium tuberculosis (Mtb). The results showed that Mtb DNA was necessary for maximal joint inflammation in adjuvant arthritis but could be replaced by synthetic ISS oligodeoxynucleotides. The arthritis-promoting effect of the Mtb DNA or of the ISS oligodeoxynucleotides correlated with an increased Th1 response to Mtb Ags, as measured by the production of IFN-γ and increased production of the osteoclast differentiation factor, receptor activator of NF-κB ligand (RANKL). The Mtb DNA did not enter the joints but dispersed to the bone marrow and spleen before the onset of systemic joint inflammation. Thus, adjuvant arthritis is a microbial DNA-dependent disease. In this model, we postulate that massive and prolonged activation of macrophages, dendritic cells, and osteoclast precursors in the bone marrow may prime the joints for the induction of inflammatory Th1 immune responses to Mtb Ags.


Blood | 2008

Autologous bone marrow transplantation in autoimmune arthritis restores immune homeostasis through CD4+ CD25+ Foxp3+ regulatory T cells.

Sarah Roord; W de Jager; Louis Boon; N Wulffraat; Anton Martens; A. B. J. Prakken; F. van Wijk

Despite the earlier use of potent immunosuppressive or cytostatic drugs and the recent emergence of biologicals as treatment for human autoimmune diseases (AIDs), some patients still remain unresponsive to treatment. To those severely ill patients, autologous bone marrow transplantation (aBMT) is applied as a last resource, leading to disease remission in a majority of patients. The underlying mechanism of action of aBMT is still largely unknown. Here, we showed that regulatory T cells (Tregs) play a role in the natural disease course of proteoglycan-induced arthritis (PGIA) and in disease remission by aBMT. aBMT led to an initial phase of rapid disease improvement corresponding with a relative increase in CD4(+)CD25(+) T cells. At this time, the CD4(+)CD25(+) cells did not yet show an increase in Foxp3 expression and showed less potent suppression. After this initial improvement, disease relapsed but stabilized at a level below the severity before aBMT. This second phase was actively regulated by potently suppressive CD4(+)CD25(+)Foxp3(+) Tregs. This work provided further insight into the role of Tregs in restoration of the immune balance by aBMT and can open the way to explore therapeutic interventions to further improve treatment of AID and disease relapses.


Springer Seminars in Immunopathology | 2003

Heat shock protein 60 and adjuvant arthritis: a model for T cell regulation in human arthritis

Berent J. Prakken; Sarah Roord; Arash Ronaghy; Marca H. M. Wauben; Salvatore Albani; Willem van Eden

Abstract. Heat shock proteins (hsp) are highly conserved, immune-dominant microbial proteins, whose expression is increased at sites of inflammation. In the experimental model of adjuvant arthritis (AA) immune responses to hsp determine the outcome of disease. AA can be transferred with a single T cell clone specific for a sequence of mycobacterial hsp65 (Mhsp65). Immunization with whole Mhsp65 on the other hand, protects in virtually all forms of experimental arthritis, including AA. This protective effect seems the consequence of the induction of a T cell response directed against self-hsp60. A similar protective effect of self-hsp60-specific T cells seems present in patients with a spontaneous remitting form of juvenile idiopathic arthritis. Next to hsp60, other hsp have similar protective effects in arthritis, while other conserved microbial proteins lack such capacity. Nasal administration of hsp60 peptides induces IL-10-driven regulatory T cells that are highly effective in suppressing arthritis. Thus hsp60, or peptides derived from hsp60, are suitable candidates for immune therapy in chronic arthritis.


PLOS ONE | 2006

Modulation of T cell function by combination of epitope specific and low dose anticytokine therapy controls autoimmune arthritis.

Sarah Roord; Evelien Zonneveld-Huijssoon; Tho Le; Gisella Puga Yung; Eva Koffeman; Arash Ronaghy; Negar Ghahramani; Paola Lanza; Rosario Billetta; Berent J. Prakken; Salvatore Albani

Innate and adaptive immunity contribute to the pathogenesis of autoimmune arthritis by generating and maintaining inflammation, which leads to tissue damage. Current biological therapies target innate immunity, eminently by interfering with single pro-inflammatory cytokine pathways. This approach has shown excellent efficacy in a good proportion of patients with Rheumatoid Arthritis (RA), but is limited by cost and side effects. Adaptive immunity, particularly T cells with a regulatory function, plays a fundamental role in controlling inflammation in physiologic conditions. A growing body of evidence suggests that modulation of T cell function is impaired in autoimmunity. Restoration of such function could be of significant therapeutic value. We have recently demonstrated that epitope-specific therapy can restore modulation of T cell function in RA patients. Here, we tested the hypothesis that a combination of anti-cytokine and epitope-specific immunotherapy may facilitate the control of autoimmune inflammation by generating active T cell regulation. This novel combination of mucosal tolerization to a pathogenic T cell epitope and single low dose anti-TNFα was as therapeutically effective as full dose anti-TNFα treatment. Analysis of the underlying immunological mechanisms showed induction of T cell immune deviation.


Annals of the Rheumatic Diseases | 2002

Balancing the immune system: Th1 and Th2

W. van Eden; R. van der Zee; P.J.S. van Kooten; Suzanne E. Berlo; Pieter M. Cobelens; Annemieke Kavelaars; Cobi J. Heijnen; Berent J. Prakken; Sarah Roord; Salvatore Albani

CD4+ T cells are subdivided into Th1 and Th2 cells. Their relative presence or activation is thought to have a regulatory effect on immune behaviour. Until recently, the relative suppression of Th1 cells by the relative increase of Th2 activities, was thought to be a main mechanism of keeping or restoring the balance in a diseased immune system. Nowadays, however, a specialised subset of regulatory T cells is held to be responsible for the main effects of securing a balanced immune system. It is possible that heat shock proteins (hsps) are relevant antigens driving such regulation. Heat shock proteins are known to be immunodominant antigens of bacteria. They are evolutionarily strongly conserved proteins present in all eukaryotic and prokaryotic cellular organisms and are upregulated by several forms of stress. Despite (the paradigm of) self tolerance, hsp-epitopes homologous to endogenous host hsp sequences have been implicated as T cell epitopes to endow cross reactive, hsp specific T cells with the capacity to regulate inflammation, such as in experimentally induced autoimmune diseases. Such T cells were found to produce regulatory cytokines like IL10, in contrast with T cells induced with other conserved microbial proteins that are not upregulated by stress. Hsps have been implicated in immune regulation not only as upregulated targets of adaptive immunity during inflammatory stress, but recently also as triggering factors for innate immunity through activation via Toll-like receptors (TLRs). Th1 cells or proinflammatory T cells are known to produce cytokines with proinflammatory activities. Therefore they are supposed to be critically involved in inflammatory conditions such as autoimmune arthritis. Th2 or helper T cells are known to produce cytokines that help B cells to become activated and to switch their class of antibody. Some of the cytokines produced by Th2 cells (IL4, IL5, IL10, and IL13) also have immune …


Annals of the Rheumatic Diseases | 2011

Bystander suppression of experimental arthritis by nasal administration of a heat shock protein peptide

Evelien Zonneveld-Huijssoon; Sarah Roord; Wilco de Jager; Mark Klein; Salvatore Albani; Stephen M. Anderton; Wietse Kuis; Femke van Wijk; Berent J. Prakken

Objectives Mucosal immune therapy with disease-inducing antigens is an effective way to prevent experimental arthritis, but in humans these antigens are unknown. In juvenile idiopathic arthritis, however, T cell recognition of a so-called bystander antigen, heat shock protein 60 (HSP60), is associated with a good prognosis. Recently epitopes derived from HSP60, a microbial peptide (p1) and its self-homologue (p2) were reported to induce tolerogenic T cell responses in vitro in patients with arthritis. A study was undertaken to determine whether mucosal administration of these bystander epitopes can be similarly effective in suppressing arthritis. Methods Rats were treated nasally with p1, p2 or phosphate-buffered saline before arthritis induction. Arthritis scores were assessed and peptide-specific proliferative responses, phenotypic analysis, cytokine production and in vitro suppressive capacity of cells were measured in lymph nodes and spleens. CD4 spleen T cells from p1- or p2-treated rats were adoptively transferred into naïve rats that were subsequently injected with complete Freunds adjuvant for arthritis induction. Results Nasal administration of p1 prevented experimental arthritis whereas treatment with the self-homologue p2 did not. Adoptive transfer of CD4 T cells protected against experimental arthritis. Treatment with p1 increased peptide-specific and self-crossreactive interferon γ (IFNγ) production. Tumour necrosis factor α (TNFα) levels were reduced at the site of inflammation. Forkhead box P3 (FoxP3) expression remained stable but the suppressive capacity of T regulatory cells in p1-treated rats was enhanced. Conclusion p1 immune therapy induces a population of CD4 T cells with reduced TNFα and increased peptide-specific IFNγ production at the site of inflammation. This population expresses FoxP3 and has potent suppressive capacity which, upon transfer, protects against arthritis. The bystander epitope p1 may therefore be a suitable candidate for antigen-specific immunotherapy in arthritis.


Autoimmunity | 2008

Regulatory T cells in autologous stem cell transplantation for autoimmune disease

F. van Wijk; Sarah Roord; Bas Vastert; I. M. de Kleer; N Wulffraat; Berent J. Prakken

Since a decade autologous stem cell transplantation (ASCT) is successfully performed to treat patients with severe autoimmune disease. However, the mechanism of action of this intervention remains largely unknown. Scarce data from animal studies and human clinical trials indicate that, besides extensive immune ablation, restoration of regulatory immune networks is of critical importance. This review focuses on the role of naturally occurring and induced regulatory T cells in controlling immune reconstitution and restoration of immune tolerance and in preventing relapses of disease following ASCT.


Annals of the Rheumatic Diseases | 2012

TLR9 agonist CpG enhances protective nasal HSP60 peptide vaccine efficacy in experimental autoimmune arthritis

Evelien Zonneveld-Huijssoon; Femke van Wijk; Sarah Roord; Eveline M. Delemarre; Jenny Meerding; Wilco de Jager; Mark Klein; Eyal Raz; Salvatore Albani; Wietse Kuis; Marianne Boes; Berent J. Prakken

Objectives Peptide-based immune tolerance induction is considered an attractive treatment option for autoimmune diseases. The authors have developed a novel method that can enhance the induction of protective peptide-specific T-cell responses, using a rat arthritis model. The authors focused on the Toll-like receptor 9 ligand CpG, which was shown to stimulate regulatory T-cell proliferation when added to plasmacytoid dendritic cells (pDC) using in-vitro cultures. Methods The peptide used is a heat shock protein 60 epitope (p1) that elicits tolerogenic peptide-specific immune responses in human arthritis patients and was recently shown to have protective capacity as a bystander antigen in the rat adjuvant arthritis model. Rats were treated with three nasal doses of p1, CpG or a combination of p1 and CpG. Antigen-presenting cells were studied in nose-draining lymph nodes (mandibular lymph nodes; MLN) after nasal treatment, and T-cell responses were analysed in joint-draining lymph nodes after arthritis induction. Results Nasal co-administration of p1/CpG significantly augmented the arthritis-protective effect of p1, while CpG treatment alone did not. Co-treatment of p1/CpG increased both the number and activation status of pDC in draining MLN, which was accompanied by amplified p1-specific T-cell proliferation and interleukin (IL)-10 production. During early arthritis, p1-specific IL-10 production was identified at the site of inflammation. P1 and p1/CpG-treated rats showed a greater amount of CD4+FoxP3+ regulatory T cells in the joint-draining lymph nodes, which correlated with lower arthritis scores. Conclusions These clinical and immunological data suggest the use of CpG as a potent adjuvant for mucosal peptide-specific immune therapy in arthritis.


Current Stem Cell Research & Therapy | 2011

Restoration of the Immune Balance by Autologous Bone Marrow Transplantation in Juvenile Idiopathic Arthritis

Eveline M. Delemarre; Sarah Roord; Nico Wulffraat; Femke van Wijk; Berent J. Prakken

Juvenile idiopathic arthritis (JIA) is one of the most frequent autoimmune diseases in childhood and is characterized by chronic inflammation of the synovial fluid in joints. Several drugs are available for the treatment of JIA, including various biological agents that interfere with critical cytokine pathways. Though very effective in suppressing disease activity, none of these drugs can cure the disease and induce a lasting medication free remission. A small proportion of JIA patients will become or are unresponsive to any form of medical treatment. For these severely ill patients autologous bone marrow transplantation (aBMT) is a last resort treatment. aBMT is remarkably effective in suppressing disease activity, with beneficial outcome reported in around 70% of these previously refractory patients. Moreover aBMT is the only treatment that can induce a lasting medication-free-disease remission in these patients. In the very long term (after 7 years of remission) however, some disease relapses are observed, with the disease returning in a less severe form compared to prior aBMT. The exact mechanism of how aBMT is inducing this lasting disease remission is still largely unknown, but data from both animal models and humans suggest a prominent role for regulatory T cells. In this review we reviewed the current views of the cellular mechanisms that lay beneath disease induction of JIA and the disease remission caused by aBMT therapy.

Collaboration


Dive into the Sarah Roord's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Salvatore Albani

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anton Martens

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arash Ronaghy

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Berent Prakken

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge