Network


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

Hotspot


Dive into the research topics where Bart J. L. Eggen is active.

Publication


Featured researches published by Bart J. L. Eggen.


Glia | 2012

Identification of a microglia phenotype supportive of remyelination

Marta Olah; Sandra Amor; Nieske Brouwer; Jonathan Vinet; Bart J. L. Eggen; Knut Biber; Hendrikus Boddeke

In multiple sclerosis, endogenous oligodendrocyte precursor cells (OPCs) attempt to remyelinate areas of myelin damage. During disease progression, however, these attempts fail. It has been suggested that modulating the inflammatory environment of the lesion might provide a promising therapeutic approach to promote endogenous remyelination. Microglia are known to play a central role in neuroinflammatory processes. To investigate the microglia phenotype that supports remyelination, we performed genome‐wide gene expression analysis of microglia from the corpus callosum during demyelination and remyelination in the mouse cuprizone model, in which remyelination spontaneously occurs after an episode of toxin‐induced primary demyelination. We provide evidence for the existence of a microglia phenotype that supports remyelination already at the onset of demyelination and persists throughout the remyelination process. Our data show that microglia are involved in the phagocytosis of myelin debris and apoptotic cells during demyelination. Furthermore, they express a cytokine and chemokine repertoire enabling them to activate and recruit endogenous OPCs to the lesion site and deliver trophic support during remyelination. This study not only provides a detailed transcriptomic analysis of the remyelination‐supportive microglia phenotype but also reinforces the notion that the primary function of microglia is the maintenance of tissue homeostasis and the support of regeneration already at the earliest stages in the development of demyelinating lesions.


Behavioral Neuroscience | 2007

Exercise Improves Memory Acquisition and Retrieval in the Y-Maze Task: Relationship With Hippocampal Neurogenesis

K. van der Borght; Robbert Havekes; T. Bos; Bart J. L. Eggen; van der Eddy Zee

Enhanced physical activity is associated with improvements in cognitive function in rodents as well as in humans. The authors examined in detail which aspects of learning and memory are influenced by exercise, using a spatial Y-maze test combined with a 14-day exercise paradigm at different stages of learning. The authors show that 14 days of wheel running promotes memory acquisition, memory retention, and reversal learning. The exercise paradigm that was employed also significantly increased the number of maturing neurons, suggesting that an increase in neurogenesis underlies the positive effects of exercise on Y-maze performance. Finally, the authors show that memory acquisition in itself does not have a major impact on the number of immature neurons. However, memory retention testing and reversal learning both cause a significant reduction in the number of doublecortin and Ser133- phosphorylated pCREB-positive cells, indicating that a decrease in neurogenesis might be a prerequisite for optimal memory retrieval.


Hippocampus | 2009

Physical exercise leads to rapid adaptations in hippocampal vasculature: Temporal dynamics and relationship to cell proliferation and neurogenesis

Karin Van der Borght; Dóra É. Kóbor-Nyakas; Karin Klauke; Bart J. L. Eggen; Csaba Nyakas; Eddy A. Van der Zee; Peter Meerlo

Increased levels of angiogenesis and neurogenesis possibly mediate the beneficial effects of physical activity on hippocampal plasticity. This study was designed to investigate the temporal dynamics of exercise‐induced changes in hippocampal angiogenesis and cell proliferation. Mice were housed with a running wheel for 1, 3, or 10 days. Analysis of glucose transporter Glut1‐positive vessel density showed a significant increase after 3 days of wheel running. Cell proliferation in the dentate gyrus showed a trend towards an increase after 3 days of running and was significantly elevated after 10 days of physical exercise. Ten days of wheel running resulted in a near‐significant increase in the number of immature neurons, as determined by a doublecortin (DCX) staining. In the second part of the study, the persistence of the exercise‐induced changes in angiogenesis and cell proliferation was determined. The running wheel was removed from the cage after 10 days of physical activity. Glut‐1 positive vessel density and hippocampal cell proliferation were determined 1 and 6 days after removal of the wheel. Both parameters had returned to baseline 24 h after cessation of physical activity. The near‐significant increase in the number of DCX‐positive immature neurons persisted for at least 6 days, indicating that new neurons formed during the period of increased physical activity had survived. Together these experiments show that the hippocampus displays a remarkable angiogenic and neurogenic plasticity and rapidly responds to changes in physical activity.


Acta neuropathologica communications | 2015

Induction of a common microglia gene expression signature by aging and neurodegenerative conditions: a co-expression meta-analysis

Inge R. Holtman; Divya Raj; Jeremy A. Miller; Wandert Schaafsma; Zhuoran Yin; Nieske Brouwer; Paul D. Wes; Thomas Möller; Marie Orre; Willem Kamphuis; Elly M. Hol; Erik Boddeke; Bart J. L. Eggen

IntroductionMicroglia are tissue macrophages of the central nervous system that monitor brain homeostasis and react upon neuronal damage and stress. Aging and neurodegeneration induce a hypersensitive, pro-inflammatory phenotype, referred to as primed microglia. To determine the gene expression signature of priming, the transcriptomes of microglia in aging, Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS) mouse models were compared using Weighted Gene Co-expression Network Analysis (WGCNA).ResultsA highly consistent consensus transcriptional profile of up-regulated genes was identified, which prominently differed from the acute inflammatory gene network induced by lipopolysaccharide (LPS). Where the acute inflammatory network was significantly enriched for NF-κB signaling, the primed microglia profile contained key features related to phagosome, lysosome, antigen presentation, and AD signaling. In addition, specific signatures for aging, AD, and ALS were identified.ConclusionMicroglia priming induces a highly conserved transcriptional signature with aging- and disease-specific aspects.


PLOS ONE | 2015

Glioma-Associated Microglia/Macrophages Display an Expression Profile Different from M1 and M2 Polarization and Highly Express Gpnmb and Spp1

Frank Szulzewsky; Andreas Pelz; Xi Feng; Michael Synowitz; Darko Markovic; Thomas Langmann; Inge R. Holtman; Xi Wang; Bart J. L. Eggen; Hendrikus Boddeke; Dolores Hambardzumyan; Susanne A. Wolf; Helmut Kettenmann

Malignant glioma belong to the most aggressive neoplasms in humans with no successful treatment available. Patients suffering from glioblastoma multiforme (GBM), the highest-grade glioma, have an average survival time of only around one year after diagnosis. Both microglia and peripheral macrophages/monocytes accumulate within and around glioma, but fail to exert effective anti-tumor activity and even support tumor growth. Here we use microarray analysis to compare the expression profiles of glioma-associated microglia/macrophages and naive control cells. Samples were generated from CD11b+ MACS-isolated cells from naïve and GL261-implanted C57BL/6 mouse brains. Around 1000 genes were more than 2-fold up- or downregulated in glioma-associated microglia/macrophages when compared to control cells. A comparison with published data sets of M1, M2a,b,c-polarized macrophages revealed a gene expression pattern that has only partial overlap with any of the M1 or M2 gene expression patterns. Samples for the qRT-PCR validation of selected M1 and M2a,b,c-specific genes were generated from two different glioma mouse models and isolated by flow cytometry to distinguish between resident microglia and invading macrophages. We confirmed in both models the unique glioma-associated microglia/macrophage phenotype including a mixture of M1 and M2a,b,c-specific genes. To validate the expression of these genes in human we MACS-isolated CD11b+ microglia/macrophages from GBM, lower grade brain tumors and control specimens. Apart from the M1/M2 gene analysis, we demonstrate that the expression of Gpnmb and Spp1 is highly upregulated in both murine and human glioma-associated microglia/macrophages. High expression of these genes has been associated with poor prognosis in human GBM, as indicated by patient survival data linked to gene expression data. We also show that microglia/macrophages are the predominant source of these transcripts in murine and human GBM. Our findings provide new potential targets for future anti-glioma therapy.


Cancer Research | 2005

RET-familial medullary thyroid carcinoma mutants Y791F and S891A activate a Src/JAK/STAT3 pathway, independent of glial cell line-derived neurotrophic factor

Ivan Plaza Menacho; Roelof Koster; Almer M. van der Sloot; Wim J. Quax; Jan Osinga; Tineke van der Sluis; Harry Hollema; G Burzynski; Oliver Gimm; Charles H.C.M. Buys; Bart J. L. Eggen; Robert M. W. Hofstra

The RET proto-oncogene encodes a receptor tyrosine kinase whose dysfunction plays a crucial role in the development of several neural crest disorders. Distinct activating RET mutations cause multiple endocrine neoplasia type 2A (MEN2A), type 2B (MEN2B), and familial medullary thyroid carcinoma (FMTC). Despite clear correlations between the mutations found in these cancer syndromes and their phenotypes, the molecular mechanisms connecting the mutated receptor to the different disease phenotypes are far from completely understood. Luciferase reporter assays in combination with immunoprecipitations, and Western and immunohistochemistry analyses were done in order to characterize the signaling properties of two FMTC-associated RET mutations, Y791F and S891A, respectively, both affecting the tyrosine kinase domain of the receptor. We show that these RET-FMTC mutants are monomeric receptors which are autophosphorylated and activated independently of glial cell line-derived neurotrophic factor. Moreover, we show that the dysfunctional signaling properties of these mutants, when compared with wild-type RET, involve constitutive activation of signal transducers and activators of transcription 3 (STAT3). Furthermore, we show that STAT3 activation is mediated by a signaling pathway involving Src, JAK1, and JAK2, differing from STAT3 activation promoted by RET(C634R) which was previously found to be independent of Src and JAKs. Three-dimensional modeling of the RET catalytic domain suggested that the structural changes promoted by the respective amino acids substitutions lead to a more accessible substrate and ATP-binding monomeric conformation. Finally, immunohistochemical analysis of FMTC tumor samples support the in vitro data, because nuclear localized, Y705-phosphorylated STAT3, as well as a high degree of RET expression at the plasma membrane was observed.


Behavioral Neuroscience | 2005

Morris water maze learning in two rat strains increases the expression of the polysialylated form of the neural cell adhesion molecule in the dentate gyrus but has no effect on hippocampal neurogenesis

K. van der Borght; A.E. Wallinga; P.G.M. Luiten; Bart J. L. Eggen; E.A. (Eddy) van der Zee

In the current study, the authors investigated whether Morris water maze learning induces alterations in hippocampal neurogenesis or neural cell adhesion molecule (NCAM) polysialylation in the dentate gyrus. Two frequently used rat strains, Wistar and Sprague-Dawley, were trained in the spatial or the nonspatial version of the water maze. Both training paradigms did not have an effect on survival of newly formed cells that were labeled 7-9 days prior to the training or on progenitor proliferation in the subgranular zone. However, the granule cell layer of the spatially trained rats contained significantly more positive cells of the polysialylated form of the NCAM. These data demonstrate that Morris water maze learning causes plastic change in the dentate gyrus without affecting hippocampal neurogenesis.


Journal of Neuroimmune Pharmacology | 2013

Microglial Phenotype and Adaptation

Bart J. L. Eggen; Disha Raj; Uwe-Karsten Hanisch; Hendrikus Boddeke

Microglia are the prime innate immune cells of the central nervous system. They can transit from a (so-called) resting state under homeostatic conditions towards a pro-inflammatory activation state upon homeostatic disturbances. Under neurodegenerative conditions, microglia have been largely perceived as neurotoxic cells. It is now becoming clear that resting microglia are not inactive but that they serve house-keeping functions. Moreover, microglia activity is not limited to proinflammatory responses, but covers a spectrum of reactive profiles. Depending on the actual situation, activated microglia display specific effector functions supporting inflammation, tissue remodeling, synaptic plasticity and neurogenesis. Many of these functions not only relate to the current state of the local neural environment but also depend on previous experience. In this review, we address microglia functions with respect to determining factors, phenotypic presentations, adaptation to environmental signals and aging. Finally, we point out primary mechanisms of microglia activation, which may comprise therapeutic targets to control neuro-inflammatory and neurodegenerative activity.


Journal of Cell Biology | 2007

UTF1 is a chromatin-associated protein involved in ES cell differentiation

Vincent van den Boom; Susanne M. Kooistra; Marije Boesjes; Bart Geverts; Adriaan B. Houtsmuller; Koshiro Monzen; Issei Komuro; Jeroen Essers; Loes J. Drenth-Diephuis; Bart J. L. Eggen

Embryonic stem (ES) cells are able to grow indefinitely (self-renewal) and have the potential to differentiate into all adult cell types (pluripotency). The regulatory network that controls pluripotency is well characterized, whereas the molecular basis for the transition from self-renewal to the differentiation of ES cells is much less understood, although dynamic epigenetic gene silencing and chromatin compaction are clearly implicated. In this study, we report that UTF1 (undifferentiated embryonic cell transcription factor 1) is involved in ES cell differentiation. Knockdown of UTF1 in ES and carcinoma cells resulted in a substantial delay or block in differentiation. Further analysis using fluorescence recovery after photobleaching assays, subnuclear fractionations, and reporter assays revealed that UTF1 is a stably chromatin-associated transcriptional repressor protein with a dynamic behavior similar to core histones. An N-terminal Myb/SANT domain and a C-terminal domain containing a putative leucine zipper are required for these properties of UTF1. These data demonstrate that UTF1 is a strongly chromatin-associated protein involved in the initiation of ES cell differentiation.


Experimental Hematology | 2003

Erythropoietin-induced serine 727 phosphorylation of STAT3 in erythroid cells is mediated by a MEK-, ERK-, and MSK1-dependent pathway

Albertus T. J. Wierenga; Irma Vogelzang; Bart J. L. Eggen; Edo Vellenga

OBJECTIVE Erythropoietin (EPO) is a key regulator of erythropoiesis, playing a role in both the proliferation and differentiation of erythroid cells. One of the signal transduction molecules activated upon EPO stimulation is signal transducer and activator of transcription (STAT) 3. Besides tyrosine 705 phosphorylation of STAT3, serine 727 phosphorylation has been described upon EPO stimulation. In the present study, we investigated which molecular pathways mediate the STAT3 serine 727 phosphorylation and the functional implications of this phosphorylation. METHODS The EPO-dependent erythroid cell line ASE2 was used to investigate which signaling routes were involved in the STAT3 serine 727 phosphorylation. Western blotting using phosphospecific antibodies was used to assess the phosphorylation status of STAT3 molecules. Transfection analysis was performed to investigate the transactivational potential of STAT3, and quantitative RT-PCR was used to study the in vivo gene expression of STAT3-responsive genes. RESULTS Western blotting of extracts of cells exposed to various chemical inhibitors revealed that the MEK inhibitors PD98059 and U0126 abrogated the EPO-mediated STAT3 serine 727 phosphorylation without an effect on tyrosine phosphorylation. Further analysis showed that MSK1 is activated downstream of ERK, and retroviral transductions with kinase-inactive MSK1 revealed that MSK1 is necessary for STAT3 serine phosphorylation. Furthermore, the STAT3-mediated transactivation was reduced by blocking the STAT3 serine phosphorylation with the MEK inhibitor U0126 or by expression of kinase-inactive MSK1. CONCLUSIONS The EPO-induced STAT3 serine 727 phosphorylation is mediated by a pathway involving MEK, ERK, and MSK1. Furthermore, serine phosphorylation of STAT3 augments the transactivational potential of STAT3.

Collaboration


Dive into the Bart J. L. Eggen's collaboration.

Top Co-Authors

Avatar

Inge R. Holtman

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Erik Boddeke

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Nieske Brouwer

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Hendrikus Boddeke

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Edo Vellenga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Hein Schepers

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Divya Raj

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Albertus T. J. Wierenga

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Ilia D. Vainchtein

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Jon D. Laman

University Medical Center Groningen

View shared research outputs
Researchain Logo
Decentralizing Knowledge