Network


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

Hotspot


Dive into the research topics where Fredrik Ivars is active.

Publication


Featured researches published by Fredrik Ivars.


European Journal of Immunology | 2000

CD4+CD25+ regulatory T cells down‐regulate co‐stimulatory molecules on antigen‐presenting cells

Lukas Cederbom; Håkan Hall; Fredrik Ivars

CD4+CD25+ T cells have been shown to inhibit experimentally induced organ‐specific autoimmune disease and depletion of these regulatory T cells from normal mice results in development of such conditions. Furthermore, CD4+CD25+ T cells suppress the IL‐2 production and thereby the proliferation of polyclonally activated CD4+CD25– T cells in vitro. The suppression in vitro is independent of secreted factors but requires interactions between CD4+CD25– and CD4+CD25+ T cells and antigen‐presenting cells (APC). We have now further investigated the function of CD4+CD25+ T cells in vitro and have focused on their interactions with APC. We found that CD4+CD25+ T cells down‐regulated the expression of the co‐stimulatory molecules CD80 and CD86 on dendritic cells. The steady‐state level of CD80 mRNA was also decreased, while the steady‐state level of CD86 mRNA was not, suggesting that distinct mechanisms regulate the expression of these molecules. The down‐regulation occurred even in the presence of stimuli that would normally increase the expression of CD80 and CD86 molecules. Thus, down‐regulation of co‐stimulatory molecules may be an additional effector function of these regulatory T cells.


Journal of Experimental Medicine | 2006

Dystroglycan is selectively cleaved at the parenchymal basement membrane at sites of leukocyte extravasation in experimental autoimmune encephalomyelitis.

Smriti Agrawal; Per Anderson; Madeleine Durbeej; Nico van Rooijen; Fredrik Ivars; Ghislain Opdenakker; Lydia Sorokin

The endothelial cell monolayer of cerebral vessels and its basement membrane (BM) are ensheathed by the astrocyte endfeet, the leptomeningeal cells, and their associated parenchymal BM, all of which contribute to establishment of the blood–brain barrier (BBB). As a consequence of this unique structure, leukocyte penetration of cerebral vessels is a multistep event. In mouse experimental autoimmune encephalomyelitis (EAE), a widely used central nervous system inflammatory model, leukocytes first penetrate the endothelial cell monolayer and underlying BM using integrin β1-mediated processes, but mechanisms used to penetrate the second barrier defined by the parenchymal BM and glia limitans remain uninvestigated. We show here that macrophage-derived gelatinase (matrix metalloproteinase [MMP]-2 and MMP-9) activity is crucial for leukocyte penetration of the parenchymal BM. Dystroglycan, a transmembrane receptor that anchors astrocyte endfeet to the parenchymal BM via high affinity interactions with laminins 1 and 2, perlecan and agrin, is identified as a specific substrate of MMP-2 and MMP-9. Ablation of both MMP-2 and MMP-9 in double knockout mice confers resistance to EAE by inhibiting dystroglycan cleavage and preventing leukocyte infiltration. This is the first description of selective in situ proteolytic damage of a BBB-specific molecule at sites of leukocyte infiltration.


Immunology | 2006

Cytotoxic T lymphocyte antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in CD4+ CD25+ regulatory T-cell-mediated suppression.

Cecilia Oderup; Lukas Cederbom; Anna Makowska; Corrado M. Cilio; Fredrik Ivars

We have previously demonstrated that CD4+ CD25+ natural regulatory T cells (Treg cells) induce down‐modulation of CD80 and CD86 (B7) molecules on dendritic cells (DCs) in vitro. In this report we show that the extent of down‐modulation is functionally significant because Treg‐cell conditioned DCs induced poor T‐cell proliferation responses. Further, we report that down‐modulation was induced rapidly and was inhibited by blocking cytotoxic T lymphocyte antigen‐4 (CTLA‐4), which is constitutively expressed by the Treg cells. Even though Treg cells have previously been reported to kill antigen‐presenting cells, the down‐modulation was not due to selective killing of DCs expressing high level of the costimulatory molecules. We propose that Treg cells down‐modulate B7‐molecules on DCs in a CTLA‐4‐dependent way, thereby enhancing suppression of T‐cell activity.


PLOS Biology | 2009

Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides.

Per Björk; Anders Björk; Thomas Vogl; Martin Stenström; David Liberg; Anders G Olsson; J. Roth; Fredrik Ivars; Tomas Leanderson

Despite more than 25 years of research, the molecular targets of quinoline-3-carboxamides have been elusive although these compounds are currently in Phase II and III development for treatment of autoimmune/inflammatory diseases in humans. Using photoaffinity cross-linking of a radioactively labelled quinoline-3-carboxamide compound, we could determine a direct association between human S100A9 and quinoline-3-carboxamides. This interaction was strictly dependent on both Zn++ and Ca++. We also show that S100A9 in the presence of Zn++ and Ca++ is an efficient ligand of receptor for advanced glycation end products (RAGE) and also an endogenous Toll ligand in that it shows a highly specific interaction with TLR4/MD2. Both these interactions are inhibited by quinoline-3-carboxamides. A clear structure-activity relationship (SAR) emerged with regard to the binding of quinoline-3-carboxamides to S100A9, as well as these compounds potency to inhibit interactions with RAGE or TLR4/MD2. The same SAR was observed when the compounds ability to inhibit acute experimental autoimmune encephalomyelitis in mice in vivo was analysed. Quinoline-3-carboxamides would also inhibit TNFα release in a S100A9-dependent model in vivo, as would antibodies raised against the quinoline-3-carboxamide–binding domain of S100A9. Thus, S100A9 appears to be a focal molecule in the control of autoimmune disease via its interactions with proinflammatory mediators. The specific binding of quinoline-3-carboxamides to S100A9 explains the immunomodulatory activity of this class of compounds and defines S100A9 as a novel target for treatment of human autoimmune diseases.


Nature Medicine | 2009

Endothelial basement membrane laminin |[alpha]|5 selectively inhibits T lymphocyte extravasation into the brain

Chuan Wu; Fredrik Ivars; Per Anderson; Rupert Hallmann; Dietmar Vestweber; Per Nilsson; Horst Robenek; Karl Tryggvason; Jian Song; Eva Korpos; Karin Loser; Stefan Beissert; Elisabeth Georges-Labouesse; Lydia Sorokin

Specific inhibition of the entry of encephalitogenic T lymphocytes into the central nervous system in multiple sclerosis would provide a means of inhibiting disease without compromising innate immune responses. We show here that targeting lymphocyte interactions with endothelial basement membrane laminins provides such a possibility. In mouse experimental autoimmune encephalomyelitis, T lymphocyte extravasation correlates with sites expressing laminin α4 and small amounts of laminin α5. In mice lacking laminin α4, laminin α5 is ubiquitously expressed along the vascular tree, resulting in marked and selective reduction of T lymphocyte infiltration into the brain and reduced disease susceptibility and severity. Vessel phenotype and immune response were not affected in these mice. Rather, laminin α5 directly inhibited integrin α6β1–mediated migration of T lymphocytes through laminin α4. The data indicate that T lymphocytes use mechanisms distinct from other immune cells to penetrate the endothelial basement membrane barrier, permitting specific targeting of this immune cell population.


PLOS ONE | 2012

S100A9 Interaction with TLR4 Promotes Tumor Growth

Eva Källberg; Thomas Vogl; David Liberg; Anders Olsson; Per Björk; Pernilla Wikström; Anders Bergh; J. Roth; Fredrik Ivars; Tomas Leanderson

By breeding TRAMP mice with S100A9 knock-out (S100A9−/−) animals and scoring the appearance of palpable tumors we observed a delayed tumor growth in animals devoid of S100A9 expression. CD11b+ S100A9 expressing cells were not observed in normal prostate tissue from control C57BL/6 mice but were readily detected in TRAMP prostate tumors. Also, S100A9 expression was observed in association with CD68+ macrophages in biopsies from human prostate tumors. Delayed growth of TRAMP tumors was also observed in mice lacking the S100A9 ligand TLR4. In the EL-4 lymphoma model tumor growth inhibition was observed in S100A9−/− and TLR4−/−, but not in RAGE−/− animals lacking an alternative S100A9 receptor. When expression of immune-regulating genes was analyzed using RT-PCR the only common change observed in mice lacking S100A9 and TLR4 was a down-regulation of TGFβ expression in splenic CD11b+ cells. Lastly, treatment of mice with a small molecule (ABR-215050) that inhibits S100A9 binding to TLR4 inhibited EL4 tumor growth. Thus, S100A9 and TLR4 appear to be involved in promoting tumor growth in two different tumor models and pharmacological inhibition of S100A9-TLR4 interactions is a novel and promising target for anti-tumor therapies.


Immunology | 2012

Induction of NFκB responses by the S100A9 protein is TLR4-dependent.

Matteo Riva; Eva Källberg; Per Björk; Dóra Hancz; Thomas Vogl; J. Roth; Fredrik Ivars; Tomas Leanderson

Interactions between danger‐associated molecular patterns (DAMP) and pathogen‐associated molecular patterns (PAMP) and pattern recognition receptors such as Toll‐like receptors (TLRs) are critical for the regulation of the inflammatory process via activation of nuclear factor‐κB (NF‐κB) and cytokine secretion. In this report, we investigated the capacity of lipopolysaccharide (LPS) ‐free S100A9 (DAMP) protein to activate human and mouse cells compared with lipoprotein‐free LPS (PAMP). First, we showed that LPS and S100A9 were able to increase NF‐κB activity followed by increased cytokine and nitric oxide (NO) secretion both in human THP‐1 cells and in mouse bone marrow‐derived dendritic cells. Surprisingly, although S100A9 triggered a weaker cytokine response than LPS, we found that S100A9 more potently induced IκBα degradation and hence NF‐κB activation. Both the S100A9‐induced response and the LPS‐induced response were completely absent in TLR4 knockout mice, whereas it was only slightly affected in RAGE knockout mice. Also, we showed that LPS and S100A9 NF‐κB induction were strongly reduced in the presence of specific inhibitors of TLR‐signalling. Chloroquine reduced S100A9 but not LPS signalling, indicating that S100A9 may need to be internalized to be fully active as a TLR4 inducer. This was confirmed using A488‐labelled S100A9 that was internalized in THP‐1 cells, showing a raise in fluorescence after 30 min at 37°. Chloroquine treatment significantly reduced the fluorescence. In summary, our data indicate that both human and mouse S100A9 are TLR4 agonists. Importantly, S100A9 induced stronger NF‐κB activation albeit weaker cytokine secretion than LPS, suggesting that S100A9 and LPS activated NF‐κB in a qualitatively distinct manner.


Journal of Immunology | 2003

Superantigen-induced regulatory T cells display different suppressive functions in the presence or absence of natural CD4+CD25+ regulatory T cells in vivo.

Susanna Grundström; Lukas Cederbom; Anette Sundstedt; Peter Scheipers; Fredrik Ivars

Repeated exposures to both microbial and innocuous Ags in vivo have been reported to both eliminate and tolerize T cells after their initial activation and expansion. The remaining tolerant T cells have been shown to suppress the response of naive T cells in vitro. This feature is reminiscent of natural CD4+CD25+ regulatory T cells. However, it is not known whether the regulatory function of in vivo-tolerized T cells is similar to the function of natural CD4+CD25+ regulatory T cells. In this study, we demonstrate that CD4+CD25+ as well as CD4+CD25− T cells isolated from mice treated with superantigen three consecutive times to induce tolerance were functionally comparable to natural CD4+CD25+ regulatory T cells, albeit more potent. The different subpopulations of in vivo-tolerized CD4+ T cells efficiently down-modulated costimulatory molecules on dendritic cells, and their suppressive functions were strictly cell contact dependent. Importantly, we demonstrate that conventional CD4+CD25− T cells could also be induced to acquire regulatory functions by the same regimen in the absence of natural regulatory T cells in vivo, but that such regulatory cells were functionally different.


Arthritis Research & Therapy | 2011

Protein synthesis of the pro-inflammatory S100A8/A9 complex in plasmacytoid dendritic cells and cell surface S100A8/A9 on leukocyte subpopulations in systemic lupus erythematosus

Christian Lood; Martin Stenström; Helena Tydén; Birgitta Gullstrand; Eva Källberg; Tomas Leanderson; Lennart Truedsson; Gunnar Sturfelt; Fredrik Ivars; Anders Bengtsson

IntroductionSystemic lupus erythematosus (SLE) is an autoimmune disease with chronic or episodic inflammation in many different organ systems, activation of leukocytes and production of pro-inflammatory cytokines. The heterodimer of the cytosolic calcium-binding proteins S100A8 and S100A9 (S100A8/A9) is secreted by activated polymorphonuclear neutrophils (PMNs) and monocytes and serves as a serum marker for several inflammatory diseases. Furthermore, S100A8 and S100A9 have many pro-inflammatory properties such as binding to Toll-like receptor 4 (TLR4). In this study we investigated if aberrant cell surface S100A8/A9 could be seen in SLE and if plasmacytoid dendritic cells (pDCs) could synthesize S100A8/A9.MethodsFlow cytometry, confocal microscopy and real-time PCR of flow cytometry-sorted cells were used to measure cell surface S100A8/A9, intracellular S100A8/A9 and mRNA levels of S100A8 and S100A9, respectively.ResultsCell surface S100A8/A9 was detected on all leukocyte subpopulations investigated except for T cells. By confocal microscopy, real-time PCR and stimulation assays, we could demonstrate that pDCs, monocytes and PMNs could synthesize S100A8/A9. Furthermore, pDC cell surface S100A8/A9 was higher in patients with active disease as compared to patients with inactive disease. Upon immune complex stimulation, pDCs up-regulated the cell surface S100A8/A9. SLE patients had also increased serum levels of S100A8/A9.ConclusionsPatients with SLE had increased cell surface S100A8/A9, which could be important in amplification and persistence of inflammation. Importantly, pDCs were able to synthesize S100A8/A9 proteins and up-regulate the cell surface expression upon immune complex-stimulation. Thus, S100A8/A9 may be a potent target for treatment of inflammatory diseases such as SLE.


European Journal of Immunology | 1998

A TCR alpha chain transgene induces maturation of CD4- CD8- alpha beta+ T cells from gamma delta T cell precursors

Maria Fritsch; Åsa Andersson; Karin Petersson; Fredrik Ivars

The proportion of CD4− CD8− double‐negative (DN) α β T cells is increased both in the thymus and in peripheral lymphoid organs of TCR α chain‐transgenic mice. In this report we have characterized this T cell population to elucidate its relationship to α β and γ δ T cells. We show that the transgenic DN cells are phenotypically similar to γ δ T cells but distinct from DN NK T cells. The precursors of DN cells have neither rearranged endogenous TCRα genes nor been negatively selected by the Mlsa antigen, suggesting that they originate from a differentiation stage before the onset of TCR α chain rearrangements and CD4/CD8 gene expression. Neither in‐frame VδDδJδ nor VγJγ rearrangements are over‐represented in this population. However, since peripheral γ δ T cells with functional TCRβ gene rearrangements have been depleted in the transgenics, we propose that the transgenic DN population, at least partially, originates from the precursors of those cells. The present data lend support to the view that maturation signals to γ δ lineage‐committed precursors can be delivered via TCR α β heterodimers.

Collaboration


Dive into the Fredrik Ivars's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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