Network


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

Hotspot


Dive into the research topics where Peter H. van der Meide is active.

Publication


Featured researches published by Peter H. van der Meide.


Journal of Neuroimmunology | 2002

Interferon-β directly influences monocyte infiltration into the central nervous system

Sarah Floris; Sigrid R. Ruuls; Anne Wierinckx; Susanne M. A. van der Pol; Ed A. Döpp; Peter H. van der Meide; Christine D. Dijkstra; Helga E. de Vries

Interferon-beta (IFN-beta) has beneficial effects on the clinical symptoms of multiple sclerosis (MS) patients, but its exact mechanism of action is yet unknown. We here suggest that IFN-beta directly modulates inflammatory events at the level of cerebral endothelium. IFN-beta treatment resulted in a marked reduction of perivascular infiltrates in acute experimental allergic encephalomyelitis (EAE), the rat model for MS, which was coupled to a major decrease in the expression of the adhesion molecules ICAM-1 and VCAM-1 on brain capillaries. In vitro, IFN-beta reduced the mRNA levels and protein expression of adhesion molecules of brain endothelial cell cultures and diminished monocyte transendothelial migration. Monocyte adhesion and subsequent migration was found to be predominantly regulated by VCAM-1. These data indicate that IFN-beta exerts direct antiinflammatory effects on brain endothelial cells thereby contributing to reduced lesion formation as observed in MS patients.


Journal of Neuroimmunology | 1993

Tumor necrosis factor-α in immune-mediated demyelination and Wallerian degeneration of the rat peripheral nervous system

Guido Stoll; S. Jung; Sebastian Jander; Peter H. van der Meide; Hans-Peter Hartung

Abstract This study reports on the immunocytochemical localization of tumor necrosis factor-alpha (TNFα) in immune-mediated demyelination and Wallerian degeneration of the rat peripheral nervous system (PNS) using teased nerve fiber preparations. In experimental autoimmune neuritis induced by active immunization (EAN) or by adoptive transfer of autoreactive T cells (AT-EAN), macrophages passing blood vessels as well as macrophages adherent to nerve fibers were TNFα-positive. Large post-phagocytic macrophages at later stages of demyelination were TNFα-negative. Intraperitoneal application of an anti-TNFα antibody to EAN rats significantly reduced the degree of inflammatory demyelination, suggesting a pathogenic role for TNFα. After nerve transection only macrophages located within degenerating nerve fibers were TNFα-positive, while those entering and leaving nerves were negative. TNFα produced by macrophages seems to bevolved in immune-mediated demyelination and non-immune myelin degradation after axotomy. While interferon-gamma (IFNγ) is present in EAN nerves and may act as a local stimulus for TNF expression, the nature of this signal in Wallerian degeneration in the absence of IFNγ is unknown.


Journal of Neuroimmunology | 1991

T cell immunity and interferon-γ secretion during experimental allergic encephalomyelitis in Lewis rats

Maha Mustafa; Per Diener; Höjeberg Bo; Peter H. van der Meide; Tomas Olsson

An immunospot assay that detects single secretory cells was used to enumerate interferon-gamma secreting cells (IFN-gamma-sc) in mononuclear cell suspensions from the central nervous system (CNS) and peripheral lymphoid organs after actively induced experimental allergic encephalomyelitis (EAE) in Lewis rats. In the CNS compartment there was a significant increase in the number of IFN-gamma-sc preceding the onset of the clinical signs of EAE. Both in rats with EAE and rats immunized with Freunds complete adjuvant (FCA) the number of IFN-gamma-sc increased in peripheral lymphoid organs, as compared to non-immunized controls. In view of the potent immunoregulatory effects of IFN-gamma, its intra-CNS secretion may play a crucial role for clinicopathological events in EAE. To study the numbers of primed T cells that in response to myelin antigens produced IFN-gamma, mononuclear cell suspensions from peripheral lymphoid organs were precultured to allow for antigen uptake, presentation and T cell triggering, followed by enumeration of IFN-gamma-sc. T cells responding to a peptide of myelin basic protein (MBP) that previously have been shown encephalitogenic in Lewis rats, appeared initially and were quantitatively dominant over the course of EAE. Later, T cell reactivities to multiple regions of MBP appeared, showing that the concept of immunodominance in EAE is non-absolute and time dependent. Splenocyte cultures from EAE rats exposed to the different antigens showed a reduced number of IFN-gamma-sc compared to cultures not exposed to antigen, suggesting an antigen-induced suppression of T cell effector molecules.


Journal of Cerebral Blood Flow and Metabolism | 2003

Interferon-beta prevents cytokine-induced neutrophil infiltration and attenuates blood-brain barrier disruption

Wouter B. Veldhuis; Sarah Floris; Peter H. van der Meide; Ine M. P. Vos; Helga E. de Vries; C. D. Dijkstra; P.R. Bär; Klaas Nicolay

Inflammation can contribute to brain injury, such as that resulting from ischemia or trauma. The authors have previously shown that the cytokine interferon-beta (IFN-β) affords protection against ischemic brain injury, which was associated with a diminished infiltration of neutrophils and a reduction in blood–brain barrier (BBB) disruption. The goal of the current study was to directly assess the effects of IFN-β on neutrophil infiltration, with the use of an in vivo assay of neutrophil infiltration with relevance to ischemic brain injury. Intrastriatal injection of recombinant rat cytokine–induced neutrophil chemoattractant-1, a member of the interleukin-8 family (1 μg in 1 μL), triggered massive infiltration of neutrophils and extensive BBB disruption 6 hours later, as measured using immunofluorescence microscopy and magnetic resonance imaging in the rat, respectively. Depleting the animals of neutrophils before interleukin-8 injection prevented BBB disruption. Treatment with IFN-β (5 × 106 U/kg) almost completely prevented neutrophil infiltration and attenuated BBB damage. Gelatinase zymography showed matrix metalloproteinase-9 expression in the ipsilateral striatum after interleukin-8 injection. Both neutrophil depletion and IFN-β treatment downregulated matrix metalloproteinase-9. IFN-β has already been approved for human use as a treatment for the chronic inflammatory disorder multiple sclerosis. The potential value of IFN-β as a treatment that can attenuate acute brain inflammation is considered.


Journal of Neuroimmunology | 1988

Rat ependyma and microglia cells express class II MHC antigens after intravenous infusion of recombinant gamma interferon.

Birte Steiniger; Peter H. van der Meide

Recombinant rat gamma-interferon was administered to Lewis rats by continuous intravenous infusion. After a 3-day administration period, at various dosages, a constant pattern of class II major histocompatibility complex (MHC) antigen induction was found in the brains and cerebella. Immunohistological double staining for class II antigens and glial fibrillary acidic protein showed that the majority of newly induced cells were microglia. The endothelium of large blood vessels and ependymal cells also expressed class II antigens. These findings demonstrate that systemically raised interferon levels can affect MHC antigen expression in the brain. Astrocytes are obviously not the primary cell type to acquire class II reactivity, and thus potential antigen-presenting capacity, in this situation.


Journal of Cerebral Blood Flow and Metabolism | 2003

Interferon-Beta Blocks Infiltration of Inflammatory Cells and Reduces Infarct Volume after Ischemic Stroke in the Rat:

Wouter B. Veldhuis; Joris W. Derksen; Sarah Floris; Peter H. van der Meide; Helga E. de Vries; Janneke Schepers; Ine M. P. Vos; C. D. Dijkstra; L. Jaap Kappelle; Klaas Nicolay; P.R. Bär

The inflammatory response that exacerbates cerebral injury after ischemia is an attractive therapeutic target: it progresses over days and strongly contributes to worsening of the neurologic outcome. The authors show that, after transient ischemic injury to the rat brain, systemic application of interferon-beta (IFN-β), a cytokine with antiinflammatory properties, attenuated the development of brain infarction. Serial magnetic resonance imaging (MRI) showed that IFN-β treatment reduced lesion volume on diffusion-weighted MRI by 70% (P < 0.01) at 1 day after stroke. IFN-β attenuated the leakage of contrast agent through the blood–brain barrier (P < 0.005), indicating a better-preserved blood–brain barrier integrity. Both control and IFN-β-treated animals showed a similar degree of relative hyperperfusion of the lesioned hemisphere, indicating that neuroprotection by IFN-β was not mediated by improved cerebral perfusion as assessed 24 hours after stroke onset. IFN-β treatment resulted in an 85% reduction (P < 0.0001) in infarct volume 3 weeks later, as determined from T2-weighted MRI and confirmed by histology. This effect was achieved even when treatment was started 6 hours after stroke onset. Quantitative immunohistochemistry at 24 hours after stroke onset showed that IFN-β almost completely prevented the infiltration of neutrophils and monocytes into the brain. Gelatinase zymography showed that this effect was associated with a decrease in matrix metalloproteinase-9 expression. In conclusion, treatment with the antiinflammatory cytokine IFN-β affords significant neuroprotection against ischemia/reperfusion injury, and within a relatively long treatment window. Because IFN-β has been approved for clinical use, it may be rapidly tested in a clinical trial for its efficacy against human stroke.


Journal of Neuroimmunology | 2001

Analysis of neural stem cells by flow cytometry: cellular differentiation modifies patterns of MHC expression.

Fiona McLaren; Clive N. Svendsen; Peter H. van der Meide; Etienne Joly

Neural stem cells are currently considered very hopeful candidates for cell replacement therapy in neurodegenerative pathologies such as Parkinsons disease. Here we show that different cell types derived from neurospheres amplified in vitro can be identified by FACS analysis relying solely on physical parameters (FSC/SSC) or autofluorescence. Additionally, after treatment with a panel of inflammatory cytokines, neurospheres and their differentiated progeny were shown to express MHC antigens which could potentially cause transplant rejection. Astrocytes expressed the highest levels of MHC. Hence removing such cells prior to transplantation could potentially optimise transplant survival.


Autoimmunity | 1992

Facial nerve transection causes expansion of myelin autoreactive T cells in regional lymph nodes and T cell homing to the facial nucleus

Tomas Olsson; Per Diener; Åke Ljungdahl; B. Höjeberg; Peter H. van der Meide; Krister Kristensson

Nervous tissue expression of immunological signal and recognition molecules, as well as lymphoid tissue immune responses after facial nerve trauma was studied in male rats of the Lewis and Brown Norway (BN) strains. In both rat strains nerve transection caused within four days the appearance of IFN-gamma-like immunoreactivity in the cytoplasm of axotomized motor neurons and an induction of MHC class I and II, and CD4 molecules on surrounding glial cells to a similar extent. T lymphocytes also infiltrated the facial nuclei ipsilateral to the axotomy in all animals. The number of autoreactive T cells in superficial cervical lymph nodes, which in response to whole myelin or peptides of myelin basic protein (MBP) secreted IFN-gamma increased markedly after axotomy. This response was more conspicuous in Lewis rats, which are susceptible to experimental allergic encephalomyelitis (EAE), than in BN rats, which are EAE resistant. A proportion of the axotomized Lewis rats also developed widespread perivascular infiltration of mononuclear cells in the CNS, reminiscent of EAE. Hypothetically, a strong expansion of myelin autoreactive IFN-gamma producing T cells secondary to nerve trauma may have immunopathological consequences in genetically predisposed individuals. It is also possible that myelin reactive T cells, whether recruited to the lesioned nerve, could have impact on macrophage function during Wallerian degeneration in the distal stump.


Journal of Neuroimmunology | 1997

Nasal administration of myelin basic protein prevents relapsing experimental autoimmune encephalomyelitis in DA rats by activating regulatory cells expressing IL-4 and TGF-β mRNA

Xue-Feng Bai; Fu-Dong Shi; Bao-Guo Xiao; Hulun Li; Peter H. van der Meide; Hans Link

This study explores nasal administration of myelin basic protein (MBP) as a potential means of inducing tolerance to relapsing experimental autoimmune encephalomyelitis (PR-EAE), an experimental multiple sclerosis (MS) model that was induced in DA rats by immunization with rat spinal cord homogenate and incomplete Freunds adjuvant. DA rats received a total dosage of 0, 6, 60, 600 micrograms/rat of bovine MBP on ten consecutive days prior to immunization. EAE with typical course was observed in control rats receiving only PBS nasally, and in rats receiving 6 micrograms/rat of MBP. Rats receiving 60 micrograms/rat of MBP developed acute EAE but no relapse during 60 days of observation post immunization (p.i.). Only one of eight rats receiving 600 micrograms/rat of MBP developed slight, transient EAE. This protection was confirmed at the histology level and was associated with decreased levels of MBP-reactive IFN-gamma secreting Th1-like spleen cells on day 13 and 60 p.i. Rats receiving 60 and 600 micrograms/rat of MBP showed decreased serum anti-MBP IgG2b antibody levels on day 60 p.i., and rats receiving 600 micrograms/rat of MBP had marginally increased anti-MBP IgG1 antibody levels in serum compared to control EAE rats. Cytokine mRNA profiles in central nervous system (CNS) and spleen mononuclear cells were evaluated. Dose-dependent reduction of TNF-alpha mRNA expression were observed both in CNS and in splenocytes. Increased IL-4 and TGF-beta mRNA expression were observed in CNS of low (6 micrograms/rat) and median (60 micrograms/rat) dose of MBP tolerized rats and in splenocytes of rats tolerized with 600 micrograms/rat of MBP. We conclude that nasal administration of MBP in DA rat prevents EAE induced by immunization with whole rat spinal cord homogenate that, besides MBP, contains multiple antigenic myelin proteins. A mechanism involving MBP-reactive regulatory cells expressing IL-4 and TGF-beta mRNA acts as part in the induction of this tolerance.


Journal of Neuroimmunology | 2001

Interleukin 10 aggravates experimental autoimmune myasthenia gravis through inducing Th2 and B cell responses to AChR.

Guang-Xian Zhang; Bao-Guo Xiao; Li-Ya Yu; Peter H. van der Meide; Hans Link

The damage of acetylcholine receptor (AChR) at neuromuscular junctions of experimental autoimmune myasthenia gravis (EAMG), an animal model of human MG, is mediated by B cells which require T cell help. The Th2 associated cytokine IL-10 suppresses production of cytokines released by Th1 cells and is considered for treatment of human autoimmune diseases. To evaluate the role of IL-10 in EAMG, rhIL-10 was administered daily to Lewis rats by the subcutaneous route starting at the day of immunization and continued for 7 weeks. IL-10 failed to abrogate EAMG at low dose (0.1 or 1 microg/day) and at the dose of 3 microg/day caused earlier onset and aggravated clinical signs of EAMG when compared to EAMG rats injected with PBS only. Although Th1 responses reflected by AChR-induced lymphocyte proliferation and levels of IFN-gamma secreting cells, as well as AChR-induced Th1 cytokine mRNA expression was suppressed, augmented IL-4 mRNA expression and AChR-specific B cell responses may play an important role in the failure of IL-10 to abrogate EAMG. This study implicates a critical precaution in planning immunotherapy of IL-10 in antibody-mediated autoimmune diseases, e.g. MG.

Collaboration


Dive into the Peter H. van der Meide's collaboration.

Top Co-Authors

Avatar

Hans Link

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hans-Gustaf Ljunggren

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Hulun Li

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar

Tomas Olsson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge