Network


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

Hotspot


Dive into the research topics where Frank Szulzewsky is active.

Publication


Featured researches published by Frank Szulzewsky.


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.


Neuro-oncology | 2013

Toll-like receptor 2 mediates microglia/brain macrophage MT1-MMP expression and glioma expansion

Katyayni Vinnakota; Feng Hu; Min-Chi Ku; Petya B. Georgieva; Frank Szulzewsky; Andreas Pohlmann; Sonia Waiczies; Helmar Waiczies; Thoralf Niendorf; Seija Lehnardt; Uwe-Karsten Hanisch; Michael Synowitz; Darko Markovic; Susanne A. Wolf; Rainer Glass; Helmut Kettenmann

BACKGROUND Glioblastomas are the most aggressive primary brain tumors in humans. Microglia/brain macrophage accumulation in and around the tumor correlates with malignancy and poor clinical prognosis of these tumors. We have previously shown that microglia promote glioma expansion through upregulation of membrane type 1 matrix metalloprotease (MT1-MMP). This upregulation depends on signaling via the Toll-like receptor (TLR) adaptor molecule myeloid differentiation primary response gene 88 (MyD88). METHODS Using in vitro, ex vivo, and in vivo techniques, we identified TLR2 as the main TLR controlling microglial MT1-MMP expression and promoting microglia-assisted glioma expansion. RESULTS The implantation of mouse GL261 glioma cells into TLR2 knockout mice resulted in significantly smaller tumors, reduced MT1-MMP expression, and enhanced survival rates compared with wild-type control mice. Tumor expansion studied in organotypic brain slices depended on both parenchymal TLR2 expression and the presence of microglia. Glioma-derived soluble factors and synthetic TLR2 specific ligands induced MT1-MMP expression in microglia from wild-type mice, but no such change in MT1-MMP gene expression was observed in microglia from TLR2 knockout mice. We also found evidence that TLR1 and TLR6 cofunction with TLR2 as heterodimers in regulating MT1-MMP expression in vitro. CONCLUSIONS Our results thus show that activation of TLR2 along with TLRs 1 and/or 6 converts microglia into a glioma supportive phenotype.


Glia | 2014

The subpopulation of microglia sensitive to neurotransmitters/neurohormones is modulated by stimulation with LPS, interferon-γ, and IL-4

Maria Pannell; Frank Szulzewsky; Vitali Matyash; Susanne A. Wolf; Helmut Kettenmann

Recently, neurotransmitters/neurohormones have been identified as factors controlling the function of microglia, the immune competent cells of the central nervous system. In this study, we compared the responsiveness of microglia to neurotransmitters/neurohormones. We freshly isolated microglia from healthy adult C57Bl/6 mice and found that only a small fraction (1–20%) responded to the application of endothelin, histamine, substance P, serotonin, galanin, somatostatin, angiotensin II, vasopressin, neurotensin, dopamine, or nicotine. In cultured microglia from neonatal and adult mice, a similarly small population of cells responded to these neurotransmitters/neurohormones. To induce a proinflammatory phenotype, we applied lipopolysaccaride (LPS) or interferon‐gamma (IFN‐γ) to the cultures for 24 h. Several of the responding populations increased; however, there was no uniform pattern when comparing adult with neonatal microglia or LPS with IFN‐γ treatment. IL‐4 as an anti‐inflammatory substance increased the histamine‐, substance P‐, and somatostatin‐sensitive populations only in microglia from adult, but not in neonatal cells. We also found that the expression of different receptors was not strongly correlated, indicating that there are many different populations of microglia with a distinct set of receptors. Our results demonstrate that microglial cells are a heterogeneous population with respect to their sensitivity to neurotransmitters/neurohormones and that they are more responsive in defined activation states. GLIA 2014;62:667–679


Genes & Development | 2017

Mutant IDH1 regulates the tumor-associated immune system in gliomas

Nduka Amankulor; Youngmi Kim; Sonali Arora; Julia Kargl; Frank Szulzewsky; Mark L. Hanke; Daciana Margineantu; Aparna Rao; Hamid Bolouri; Jeff Delrow; David M. Hockenbery; A. McGarry Houghton; Eric C. Holland

Gliomas harboring mutations in isocitrate dehydrogenase 1/2 (IDH1/2) have the CpG island methylator phenotype (CIMP) and significantly longer patient survival time than wild-type IDH1/2 (wtIDH1/2) tumors. Although there are many factors underlying the differences in survival between these two tumor types, immune-related differences in cell content are potentially important contributors. In order to investigate the role of IDH mutations in immune response, we created a syngeneic pair mouse model for mutant IDH1 (muIDH1) and wtIDH1 gliomas and demonstrated that muIDH1 mice showed many molecular and clinical similarities to muIDH1 human gliomas, including a 100-fold higher concentration of 2-hydroxygluratate (2-HG), longer survival time, and higher CpG methylation compared with wtIDH1. Also, we showed that IDH1 mutations caused down-regulation of leukocyte chemotaxis, resulting in repression of the tumor-associated immune system. Given that significant infiltration of immune cells such as macrophages, microglia, monocytes, and neutrophils is linked to poor prognosis in many cancer types, these reduced immune infiltrates in muIDH1 glioma tumors may contribute in part to the differences in aggressiveness of the two glioma types.


Glia | 2015

Altered microglial phagocytosis in GPR34-deficient mice

Julia Preissler; Antje Grosche; Vera Lede; Diana Le Duc; Katja Krügel; Vitali Matyash; Frank Szulzewsky; Sonja Kallendrusch; Kerstin Immig; Helmut Kettenmann; Ingo Bechmann; Torsten Schöneberg; Angela Schulz

GPR34 is a Gi/o protein‐coupled receptor (GPCR) of the nucleotide receptor P2Y12‐like group. This receptor is highly expressed in microglia, however, the functional relevance of GPR34 in these glial cells is unknown. Previous results suggested an impaired immune response in GPR34‐deficient mice infected with Cryptococcus neoformans. Here we show that GPR34 deficiency results in morphological changes in retinal and cortical microglia. RNA sequencing analysis of microglia revealed a number of differentially expressed transcripts involved in cell motility and phagocytosis. We found no differences in microglial motility after entorhinal cortex lesion and in response to laser lesion. However, GPR34‐deficient microglia showed reduced phagocytosis activity in both retina and acutely isolated cortical slices. Our study identifies GPR34 as an important signaling component controlling microglial function, morphology and phagocytosis. GLIA 2015;63:206–215


Glia | 2016

Human glioblastoma-associated microglia/monocytes express a distinct RNA profile compared to human control and murine samples

Frank Szulzewsky; Sonali Arora; Lot de Witte; Thomas Ulas; Darko Markovic; Joachim L. Schultze; Eric C. Holland; Michael Synowitz; Susanne A. Wolf; Helmut Kettenmann

Glioblastoma (GBM) is the most aggressive brain tumor in adults. It is strongly infiltrated by microglia and peripheral monocytes that support tumor growth. In the present study we used RNA sequencing to compare the expression profile of CD11b+ human glioblastoma‐associated microglia/monocytes (hGAMs) to CD11b+ microglia isolated from non‐tumor samples. Hierarchical clustering and principal component analysis showed a clear separation of the two sample groups and we identified 334 significantly regulated genes in hGAMs. In comparison to human control microglia hGAMs upregulated genes associated with mitotic cell cycle, cell migration, cell adhesion, and extracellular matrix organization. We validated the expression of several genes associated with extracellular matrix organization in samples of human control microglia, hGAMs, and the hGAMs‐depleted fraction via qPCR. The comparison to murine GAMs (mGAMs) showed that both cell populations share a significant fraction of upregulated transcripts compared with their respective controls. These genes were mostly related to mitotic cell cycle. However, in contrast to murine cells, human GAMs did not upregulate genes associated to immune activation. Comparison of human and murine GAMs expression data to several data sets of in vitro‐activated human macrophages and murine microglia showed that, in contrast to mGAMs, hGAMs share a smaller overlap to these data sets in general and in particular to cells activated by proinflammatory stimulation with LPS + INFγ or TNFα. Our findings provide new insights into the biology of human glioblastoma‐associated microglia/monocytes and give detailed information about the validity of murine experimental models. GLIA 2016 GLIA 2016;64:1416–1436


Circulation | 2015

Vascular Signal Transducer and Activator of Transcription-3 Promotes Angiogenesis and Neuroplasticity Long-Term After Stroke

Christian J. Hoffmann; Ulrike Harms; Andre Rex; Frank Szulzewsky; Susanne A. Wolf; Ulrike Grittner; Gisela Lättig-Tünnemann; Michael Sendtner; Helmut Kettenmann; Ulrich Dirnagl; Matthias Endres; Christoph Harms

Center for Stroke Research Berlin, Charite-Universitatsmedizin Berlin, Germany; Dept of Neurology, Charite-Universitatsmedizin Berlin, Germany; Max-Delbruck Center for Molecular Medicine, Berlin, Germany; Institute of Clinical Neurobiology, University Hospital, University of Wurzburg, Germany; Cluster of Excellence NeuroCure, Charite-Universitatsmedizin Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), Partner Site, Berlin, Germany; German Center for Cardiovascular Diseases (DZHK), Partner Site, Berlin, GermanyBackground— Poststroke angiogenesis contributes to long-term recovery after stroke. Signal transducer and activator of transcription-3 (Stat3) is a key regulator for various inflammatory signals and angiogenesis. It was the aim of this study to determine its function in poststroke outcome. Methods and Results— We generated a tamoxifen-inducible and endothelial-specific Stat3 knockout mouse model by crossbreeding Stat3floxed/KO and Tie2-CreERT2 mice. Cerebral ischemia was induced by 30 minutes of middle cerebral artery occlusion. We demonstrated that endothelial Stat3 ablation did not alter lesion size 2 days after ischemia but did worsen functional outcome at 14 days and increase lesion size at 28 days. At this late time point vascular Stat3 expression and phosphorylation were still increased in wild-type mice. Gene array analysis of a CD31-enriched cell population of the neurovascular niche showed that endothelial Stat3 ablation led to a shift toward an antiangiogenic and axon growth-inhibiting micromilieu after stroke, with an increased expression of Adamts9. Remodeling and glycosylation of the extracellular matrix and microglia proliferation were increased, whereas angiogenesis was reduced. Conclusions— Endothelial Stat3 regulates angiogenesis, axon growth, and extracellular matrix remodeling and is essential for long-term recovery after stroke. It might serve as a potent target for stroke treatment after the acute phase by fostering angiogenesis and neuroregeneration.


Circulation | 2015

Vascular Stat3 Promotes Angiogenesis and Neuroplasticity Long-Term After Stroke

Christian J. Hoffmann; Ulrike Harms; Andre Rex; Frank Szulzewsky; Susanne A. Wolf; Ulrike Grittner; Gisela Lättig-Tünnemann; Michael Sendtner; Helmut Kettenmann; Ulrich Dirnagl; Matthias Endres; Christoph Harms

Center for Stroke Research Berlin, Charite-Universitatsmedizin Berlin, Germany; Dept of Neurology, Charite-Universitatsmedizin Berlin, Germany; Max-Delbruck Center for Molecular Medicine, Berlin, Germany; Institute of Clinical Neurobiology, University Hospital, University of Wurzburg, Germany; Cluster of Excellence NeuroCure, Charite-Universitatsmedizin Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), Partner Site, Berlin, Germany; German Center for Cardiovascular Diseases (DZHK), Partner Site, Berlin, GermanyBackground— Poststroke angiogenesis contributes to long-term recovery after stroke. Signal transducer and activator of transcription-3 (Stat3) is a key regulator for various inflammatory signals and angiogenesis. It was the aim of this study to determine its function in poststroke outcome. Methods and Results— We generated a tamoxifen-inducible and endothelial-specific Stat3 knockout mouse model by crossbreeding Stat3floxed/KO and Tie2-CreERT2 mice. Cerebral ischemia was induced by 30 minutes of middle cerebral artery occlusion. We demonstrated that endothelial Stat3 ablation did not alter lesion size 2 days after ischemia but did worsen functional outcome at 14 days and increase lesion size at 28 days. At this late time point vascular Stat3 expression and phosphorylation were still increased in wild-type mice. Gene array analysis of a CD31-enriched cell population of the neurovascular niche showed that endothelial Stat3 ablation led to a shift toward an antiangiogenic and axon growth-inhibiting micromilieu after stroke, with an increased expression of Adamts9. Remodeling and glycosylation of the extracellular matrix and microglia proliferation were increased, whereas angiogenesis was reduced. Conclusions— Endothelial Stat3 regulates angiogenesis, axon growth, and extracellular matrix remodeling and is essential for long-term recovery after stroke. It might serve as a potent target for stroke treatment after the acute phase by fostering angiogenesis and neuroregeneration.


Cell Reports | 2018

A De Novo Mouse Model of C11orf95-RELA Fusion-Driven Ependymoma Identifies Driver Functions in Addition to NF-κB

Tatsuya Ozawa; Sonali Arora; Frank Szulzewsky; Gordana Juric-Sekhar; Yoshiteru Miyajima; Hamid Bolouri; Yoshie Yasui; Jason Barber; Robert Kupp; James Dalton; Terreia S. Jones; Mitsutoshi Nakada; Toshihiro Kumabe; David W. Ellison; Richard J. Gilbertson; Eric C. Holland

SUMMARY The majority of supratentorial ependymomas (ST-ependymomas) have few mutations but frequently display chromothripsis of chromosome 11q that generates a fusion between C11orf95 and RELA (RELAFUS). Neural stem cells transduced with RELAFUS ex vivo form ependymomas when implanted in the brain. These tumors display enhanced NF-κB signaling, suggesting that this aberrant signal is the principal mechanism of oncogenesis. However, it is not known whether RELAFUS is sufficient to drive de novo ependymoma tumorigenesis in the brain and, if so, whether these tumors also arise from neural stem cells. We show that RELAFUS drives ST-ependymoma formation from periventricular neural stem cells in mice and that RELAFUS-induced tumorigenesis is likely dependent on a series of cell signaling pathways in addition to NF-κB.


Glia | 2017

Genetic driver mutations define the expression signature and microenvironmental composition of high-grade gliomas

C. J. Herting; Z. Chen; K. L. Pitter; Frank Szulzewsky; I. Kaffes; M. Kaluzova; J. C. Park; P. J. Cimino; Cameron Brennan; B. Wang; Dolores Hambardzumyan

High‐grade gliomas (HGG), including glioblastomas, are characterized by invasive growth, resistance to therapy, and high inter‐ and intra‐tumoral heterogeneity. The key histological hallmarks of glioblastoma are pseudopalisading necrosis and microvascular proliferation, which allow pathologists to distinguish glioblastoma from lower‐grade gliomas. In addition to being genetically and molecularly heterogeneous, HGG are also heterogeneous with respect to the composition of their microenvironment. The question of whether this microenvironmental heterogeneity is driven by the molecular identity of the tumor remains controversial. However, this question is of utmost importance since microenvironmental, non‐neoplastic cells are key components of the most radiotherapy‐ and chemotherapy‐resistant niches of the tumor. Our work demonstrates a versatile, reliable, and reproducible adult HGG mouse model with NF1‐silencing as a driver mutation. This model shows significant differences in tumor microenvironment, expression of subtype‐specific markers, and response to standard therapy when compared to our established PDGFB‐overexpressing HGG mouse model. PDGFB‐overexpressing and NF1‐silenced murine tumors closely cluster with human proneural and mesenchymal subtypes, as well as PDGFRA‐amplified and NF1‐deleted/mutant human tumors, respectively, at both the RNA and protein expression levels. These models can be generated in fully immunocompetent mixed or C57BL/6 genetic background mice, and therefore can easily be incorporated into preclinical studies for cancer cell‐specific or immune cell‐targeting drug discovery studies.

Collaboration


Dive into the Frank Szulzewsky's collaboration.

Top Co-Authors

Avatar

Helmut Kettenmann

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Susanne A. Wolf

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Eric C. Holland

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sonali Arora

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Darko Markovic

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Vitali Matyash

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Tatsuya Ozawa

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge