50 results on '"Szulzewsky F"'
Search Results
2. The subpopulation of microglia sensitive to neurotransmitters/neurohormones is modulated by stimulation with LPS, interferon-γ, and IL-4
- Author
-
Pannell, M., Szulzewsky, F., Matyash, V., Wolf, S.A., and Kettenmann, H.
- Subjects
nervous system ,Function and Dysfunction of the Nervous System - Abstract
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-{gamma}) 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-{gamma} 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.
- Published
- 2014
3. Toll-like receptor 2 mediates microglia/brain macrophage MT1-MMP expression and glioma expansion
- Author
-
Vinnakota, K, Hu, F, Ku, MC, Georgieva, PB, Szulzewsky, F, Pohlmann, A, Waiczies, S, Waiczies, H, Niendorf, T, Lehnardt, S, Hanisch, UK, Markovic, D, Wolf, SA, Glass, R, Kettenmann, H, Synowitz, M, Vinnakota, K, Hu, F, Ku, MC, Georgieva, PB, Szulzewsky, F, Pohlmann, A, Waiczies, S, Waiczies, H, Niendorf, T, Lehnardt, S, Hanisch, UK, Markovic, D, Wolf, SA, Glass, R, Kettenmann, H, and Synowitz, M
- Published
- 2014
4. Toll-like receptor 2 mediates microglia/brain macrophage MT1-MMP expression and glioma expansion
- Author
-
Vinnakota, K., primary, Hu, F., additional, Ku, M.-C., additional, Georgieva, P. B., additional, Szulzewsky, F., additional, Pohlmann, A., additional, Waiczies, S., additional, Waiczies, H., additional, Niendorf, T., additional, Lehnardt, S., additional, Hanisch, U.-K., additional, Synowitz, M., additional, Markovic, D., additional, Wolf, S. A., additional, Glass, R., additional, and Kettenmann, H., additional
- Published
- 2013
- Full Text
- View/download PDF
5. ERK signaling promotes resistance to TRK kinase inhibition in NTRK fusion-driven glioma mouse models.
- Author
-
Schmid S, Russell ZR, Yamashita AS, West ME, Parrish AG, Walker J, Rudoy D, Yan JZ, Quist DC, Gessesse BN, Alvinez N, Hill KD, Anderson LW, Cimino PJ, Kumasaka DK, Parchment RE, Holland EC, and Szulzewsky F
- Subjects
- Animals, Mice, Humans, Drug Resistance, Neoplasm genetics, Oncogene Proteins, Fusion metabolism, Oncogene Proteins, Fusion genetics, Receptor, trkC genetics, Receptor, trkC metabolism, Receptor, trkC antagonists & inhibitors, Receptor, trkB metabolism, Receptor, trkB genetics, Glioma genetics, Glioma pathology, Glioma drug therapy, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Disease Models, Animal, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System genetics, Receptor, trkA metabolism, Receptor, trkA genetics, Receptor, trkA antagonists & inhibitors
- Abstract
Pediatric-type high-grade gliomas frequently harbor gene fusions involving receptor tyrosine kinase genes, including neurotrophic tyrosine kinase receptor (NTRK) fusions. Clinically, these tumors show high initial response rates to tyrosine kinase inhibition but ultimately recur due to the accumulation of additional resistance-conferring mutations. Here, we develop a series of genetically engineered mouse models of treatment-naive and -experienced NTRK1/2/3 fusion-driven gliomas. All tested NTRK fusions are oncogenic in vivo. The NTRK variant, N-terminal fusion partners, and resistance-associated point mutations all influence tumor histology and aggressiveness. Additional tumor suppressor losses greatly enhance tumor aggressiveness. Treatment with TRK kinase inhibitors significantly extends the survival of NTRK fusion-driven glioma mice, but fails to fully eradicate tumors, leading to recurrence upon treatment discontinuation. Finally, we show that ERK activation promotes resistance to TRK kinase inhibition and identify MEK inhibition as a potential combination therapy. These models will be invaluable tools to study therapy resistance of NTRK fusion tumors., Competing Interests: Declaration of interests The authors declare that they have no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. TRKing down drug resistance in NTRK fusion-positive cancers † .
- Author
-
Parrish AG and Szulzewsky F
- Subjects
- Humans, Neurofibromin 2 genetics, Oncogene Proteins, Fusion genetics, Benzamides therapeutic use, Benzamides pharmacology, Receptor, trkA genetics, Receptor, trkA metabolism, Signal Transduction genetics, Indazoles therapeutic use, Indazoles pharmacology, Mutation, Sarcoma genetics, Sarcoma drug therapy, Sarcoma pathology, Antineoplastic Agents therapeutic use, Antineoplastic Agents pharmacology, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Drug Resistance, Neoplasm genetics, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology
- Abstract
In a recent issue of The Journal of Pathology, Chen and colleagues established novel patient-derived ex vivo models of NTRK fusion-positive soft tissue sarcoma to characterize resistance mechanisms against targeted therapy with tyrosine kinase inhibitors. Prolonged exposure to escalating concentrations of the tyrosine kinase inhibitor, entrectinib, ultimately led to the occurrence of resistant clones that harbored an inactivating mutation in the NF2 gene, not previously described in this context, accompanied by increased PI3K/AKT/mTOR and Ras/Raf/MEK/ERK signaling. Finally, an inhibitor screen identified, among others, MEK and mTOR inhibitors as potential combination agents. © 2024 The Pathological Society of Great Britain and Ireland., (© 2024 The Pathological Society of Great Britain and Ireland.)
- Published
- 2024
- Full Text
- View/download PDF
7. Aggressive high-grade NF2 mutant meningiomas downregulate oncogenic YAP signaling via the upregulation of VGLL4 and FAT3/4.
- Author
-
Parrish AG, Arora S, Thirimanne HN, Rudoy D, Schmid S, Sievers P, Sahm F, Holland EC, and Szulzewsky F
- Abstract
Background: Meningiomas are the most common primary central nervous system tumors in adults. Although generally benign, a subset is of higher grade and ultimately fatal. Around half of all meningiomas harbor inactivating mutations in NF2, leading to deregulation of oncogenic YAP1 activity. While benign NF2 mutant meningiomas exhibit few genetic events in addition to NF2 inactivation, aggressive high-grade NF2 mutant meningiomas frequently harbor a highly aberrant genome. It is unclear if NF2 mutant meningiomas of different grades are equally reliant on YAP activity., Methods: We analyzed bulk and single-cell RNA-Seq data from a large cohort of human meningiomas for the expression of YAP1 target genes and Hippo effectors as well as in vitro cell line experiments., Results: Aggressive NF2 mutant meningiomas harbor decreased expression levels of YAP1 target genes and increased expression levels of the YAP1 antagonist VGLL4 and the upstream regulators FAT3/4 compared to their benign counterparts. Decreased expression of YAP1 target genes as well as high expression of VGLL4 and FAT3/4 is significantly associated with an increased risk of recurrence. In vitro, overexpression of VGLL4 resulted in the downregulation of YAP activity in benign NF2 mutant meningioma cells, confirming the direct link between VGLL4 expression and decreased levels of YAP activity observed in aggressive NF2 mutant meningiomas., Conclusions: Our results shed new insight into the biology of benign and aggressive NF2 mutant meningiomas and may have important implications for the efficacy of therapies targeting oncogenic YAP1 activity in NF2 mutant meningiomas., Competing Interests: The authors declare that they have no competing interests., (© The Author(s) 2024. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
8. Meningioma transcriptomic landscape demonstrates novel subtypes with regional associated biology and patient outcome.
- Author
-
Thirimanne HN, Almiron-Bonnin D, Nuechterlein N, Arora S, Jensen M, Parada CA, Qiu C, Szulzewsky F, English CW, Chen WC, Sievers P, Nassiri F, Wang JZ, Klisch TJ, Aldape KD, Patel AJ, Cimino PJ, Zadeh G, Sahm F, Raleigh DR, Shendure J, Ferreira M, and Holland EC
- Subjects
- Humans, Male, Female, Middle Aged, Gene Expression Regulation, Neoplastic, Algorithms, Gene Expression Profiling methods, Meningioma genetics, Meningioma pathology, Transcriptome, Meningeal Neoplasms genetics, Meningeal Neoplasms pathology
- Abstract
Meningiomas, although mostly benign, can be recurrent and fatal. World Health Organization (WHO) grading of the tumor does not always identify high-risk meningioma, and better characterizations of their aggressive biology are needed. To approach this problem, we combined 13 bulk RNA sequencing (RNA-seq) datasets to create a dimension-reduced reference landscape of 1,298 meningiomas. The clinical and genomic metadata effectively correlated with landscape regions, which led to the identification of meningioma subtypes with specific biological signatures. The time to recurrence also correlated with the map location. Further, we developed an algorithm that maps new patients onto this landscape, where the nearest neighbors predict outcome. This study highlights the utility of combining bulk transcriptomic datasets to visualize the complexity of tumor populations. Further, we provide an interactive tool for understanding the disease and predicting patient outcomes. This resource is accessible via the online tool Oncoscape, where the scientific community can explore the meningioma landscape., Competing Interests: Declaration of interests Although the majority of Oncoscape is open source, a subset of the technology and computational algorithms presented in this paper are covered by serial no. 63/595,717, and N.N., S.A., M.J., and E.C.H. are listed as inventors., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. RNA Polymerase II hypertranscription in cancer FFPE samples.
- Author
-
Henikoff S, Henikoff JG, Paranal RM, Greene JE, Zheng Y, Russell ZR, Szulzewsky F, Kugel S, Holland EC, and Ahmad K
- Abstract
Hypertranscription is common in human cancers and predicts poor prognosis. However detection of hypertranscription is indirect, relying on accurately quantifying mRNA levels and estimating cell numbers. Previously, we introduced FFPE-CUTAC, a genome-wide method for mapping RNA Polymerase II (RNAPII) in formalin-fixed paraffin-embedded (FFPE) sections. Here we use FFPE-CUTAC to demonstrate genome-wide hypertranscription both in transgene-driven mouse gliomas and in assorted human tumors at active regulatory elements and replication-coupled histone genes with reduced mitochondrial DNA abundance. FFPE-CUTAC identified RNAPII-bound regulatory elements shared among diverse cancers and readily categorized human tumors despite using very small samples and low sequencing depths. Remarkably, RNAPII FFPE-CUTAC identified de novo and precisely mapped HER2 amplifications punctuated by likely selective sweeps including genes encoding direct positive regulators of RNAPII itself. Our results demonstrate that FFPE-CUTAC measurements of hypertranscription and classifications of tumors using small sections provides an affordable and sensitive genome-wide strategy for personalized medicine., Competing Interests: Declaration of interests S.H. is an inventor in a USPTO patent application filed by the Fred Hutchinson Cancer Center pertaining to CUTAC and FFPE-CUTAC (application number 63/505,964). The remaining authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
10. Haploinsufficiency of phosphodiesterase 10A activates PI3K/AKT signaling independent of PTEN to induce an aggressive glioma phenotype.
- Author
-
Nuechterlein N, Shelbourn A, Szulzewsky F, Arora S, Casad M, Pattwell S, Merino-Galan L, Sulman E, Arowa S, Alvinez N, Jung M, Brown D, Tang K, Jackson S, Stoica S, Chittaboina P, Banasavadi-Siddegowda YK, Wirsching HG, Stella N, Shapiro L, Paddison P, Patel AP, Gilbert MR, Abdullaev Z, Aldape K, Pratt D, Holland EC, and Cimino PJ
- Subjects
- Humans, Animals, Mice, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Haploinsufficiency, PTEN Phosphohydrolase genetics, Phosphoric Diester Hydrolases genetics, Cell Line, Tumor, Glioblastoma genetics, Glioma genetics, Brain Neoplasms genetics
- Abstract
Glioblastoma is universally fatal and characterized by frequent chromosomal copy number alterations harboring oncogenes and tumor suppressors. In this study, we analyzed exome-wide human glioblastoma copy number data and found that cytoband 6q27 is an independent poor prognostic marker in multiple data sets. We then combined CRISPR-Cas9 data, human spatial transcriptomic data, and human and mouse RNA sequencing data to nominate PDE10A as a potential haploinsufficient tumor suppressor in the 6q27 region. Mouse glioblastoma modeling using the RCAS/tv-a system confirmed that Pde10a suppression induced an aggressive glioma phenotype in vivo and resistance to temozolomide and radiation therapy in vitro. Cell culture analysis showed that decreased Pde10a expression led to increased PI3K/AKT signaling in a Pten-independent manner, a response blocked by selective PI3K inhibitors. Single-nucleus RNA sequencing from our mouse gliomas in vivo, in combination with cell culture validation, further showed that Pde10a suppression was associated with a proneural-to-mesenchymal transition that exhibited increased cell adhesion and decreased cell migration. Our results indicate that glioblastoma patients harboring PDE10A loss have worse outcomes and potentially increased sensitivity to PI3K inhibition., (© 2024 Nuechterlein et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2024
- Full Text
- View/download PDF
11. Meningioma: current updates on genetics, classification, and mouse modeling.
- Author
-
Szulzewsky F, Thirimanne HN, and Holland EC
- Subjects
- Adult, Humans, Animals, Mice, Kruppel-Like Factor 4, Mutation, Prognosis, Meningioma genetics, Meningioma pathology, Meningeal Neoplasms genetics, Meningeal Neoplasms pathology, Meningeal Neoplasms surgery
- Abstract
Meningiomas, the most common primary brain tumors in adults, are often benign and curable by surgical resection. However, a subset is of higher grade, shows aggressive growth behavior as well as brain invasion, and often recurs even after several rounds of surgery. Increasing evidence suggests that tumor classification and grading primarily based on histopathology do not always accurately predict tumor aggressiveness and recurrence behavior. The underlying biology of aggressive treatment-resistant meningiomas and the impact of specific genetic aberrations present in these high-grade tumors is still only insufficiently understood. Therefore, an in-depth research into the biology of this tumor type is warranted. More recent studies based on large-scale molecular data such as whole exome/genome sequencing, DNA methylation sequencing, and RNA sequencing have provided new insights into the biology of meningiomas and have revealed new risk factors and prognostic subtypes. The most common genetic aberration in meningiomas is functional loss of NF2 and occurs in both low- and high-grade meningiomas, whereas NF2-wildtype meningiomas are enriched for recurrent mutations in TRAF7, KLF4, AKT1, PI3KCA, and SMO and are more frequently benign. Most meningioma mouse models are based on patient-derived xenografts and only recently have new genetically engineered mouse models of meningioma been developed that will aid in the systematic evaluation of specific mutations found in meningioma and their impact on tumor behavior. In this article, we review recent advances in the understanding of meningioma biology and classification and highlight the most common genetic mutations, as well as discuss new genetically engineered mouse models of meningioma., Competing Interests: The authors have no conflicts of interest to disclose., (© 2024 The Author(s). Published by Upsala Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
12. Phase separation of YAP-MAML2 differentially regulates the transcriptome.
- Author
-
Chung CI, Yang J, Yang X, Liu H, Ma Z, Szulzewsky F, Holland EC, Shen Y, and Shu X
- Subjects
- Transcription Factors metabolism, Adaptor Proteins, Signal Transducing metabolism, Oncogenes, Transcriptome, Phase Separation
- Abstract
Phase separation (PS) drives the formation of biomolecular condensates that are emerging biological structures involved in diverse cellular processes. Recent studies have unveiled PS-induced formation of several transcriptional factor (TF) condensates that are transcriptionally active, but how strongly PS promotes gene activation remains unclear. Here, we show that the oncogenic TF fusion Yes-associated protein 1-Mastermind like transcriptional coactivator 2 (YAP-MAML2) undergoes PS and forms liquid-like condensates that bear the hallmarks of transcriptional activity. Furthermore, we examined the contribution of PS to YAP-MAML2-mediated gene expression by developing a chemogenetic tool that dissolves TF condensates, allowing us to compare phase-separated and non-phase-separated conditions at identical YAP-MAML2 protein levels. We found that a small fraction of YAP-MAML2-regulated genes is further affected by PS, which include the canonical YAP target genes CTGF and CYR61 , and other oncogenes. On the other hand, majority of YAP-MAML2-regulated genes are not affected by PS, highlighting that transcription can be activated effectively by diffuse complexes of TFs with the transcriptional machinery. Our work opens new directions in understanding the role of PS in selective modulation of gene expression, suggesting differential roles of PS in biological processes., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
13. A paracrine circuit of IL-1β/IL-1R1 between myeloid and tumor cells drives genotype-dependent glioblastoma progression.
- Author
-
Chen Z, Giotti B, Kaluzova M, Vallcorba MP, Rawat K, Price G, Herting CJ, Pinero G, Cristea S, Ross JL, Ackley J, Maximov V, Szulzewsky F, Thomason W, Marquez-Ropero M, Angione A, Nichols N, Tsankova NM, Michor F, Shayakhmetov DM, Gutmann DH, Tsankov AM, and Hambardzumyan D
- Subjects
- Animals, Humans, Mice, Genotype, Macrophages metabolism, NF-kappa B genetics, NF-kappa B metabolism, Proto-Oncogene Proteins c-sis metabolism, Receptors, Interleukin-1 metabolism, Paracrine Communication, Glioblastoma metabolism, Glioblastoma pathology, Interleukin-1beta metabolism, Receptors, Interleukin-1 Type I metabolism
- Abstract
Monocytes and monocyte-derived macrophages (MDMs) from blood circulation infiltrate glioblastoma (GBM) and promote growth. Here, we show that PDGFB-driven GBM cells induce the expression of the potent proinflammatory cytokine IL-1β in MDM, which engages IL-1R1 in tumor cells, activates the NF-κB pathway, and subsequently leads to induction of monocyte chemoattractant proteins (MCPs). Thus, a feedforward paracrine circuit of IL-1β/IL-1R1 between tumors and MDM creates an interdependence driving PDGFB-driven GBM progression. Genetic loss or locally antagonizing IL-1β/IL-1R1 leads to reduced MDM infiltration, diminished tumor growth, and reduced exhausted CD8+ T cells and thereby extends the survival of tumor-bearing mice. In contrast to IL-1β, IL-1α exhibits antitumor effects. Genetic deletion of Il1a/b is associated with decreased recruitment of lymphoid cells and loss-of-interferon signaling in various immune populations and subsets of malignant cells and is associated with decreased survival time of PDGFB-driven tumor-bearing mice. In contrast to PDGFB-driven GBM, Nf1-silenced tumors have a constitutively active NF-κB pathway, which drives the expression of MCPs to recruit monocytes into tumors. These results indicate local antagonism of IL-1β could be considered as an effective therapy specifically for proneural GBM.
- Published
- 2023
- Full Text
- View/download PDF
14. Epigenomic analysis of formalin-fixed paraffin-embedded samples by CUT&Tag.
- Author
-
Henikoff S, Henikoff JG, Ahmad K, Paranal RM, Janssens DH, Russell ZR, Szulzewsky F, Kugel S, and Holland EC
- Subjects
- Animals, Mice, Paraffin Embedding, Retrospective Studies, Formaldehyde, Epigenomics, Chromatin genetics
- Abstract
For more than a century, formalin-fixed paraffin-embedded (FFPE) sample preparation has been the preferred method for long-term preservation of biological material. However, the use of FFPE samples for epigenomic studies has been difficult because of chromatin damage from long exposure to high concentrations of formaldehyde. Previously, we introduced Cleavage Under Targeted Accessible Chromatin (CUTAC), an antibody-targeted chromatin accessibility mapping protocol based on CUT&Tag. Here we show that simple modifications of our CUTAC protocol either in single tubes or directly on slides produce high-resolution maps of paused RNA Polymerase II at enhancers and promoters using FFPE samples. We find that transcriptional regulatory element differences produced by FFPE-CUTAC distinguish between mouse brain tumors and identify and map regulatory element markers with high confidence and precision, including microRNAs not detectable by RNA-seq. Our simple workflows make possible affordable epigenomic profiling of archived biological samples for biomarker identification, clinical applications and retrospective studies., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
15. Visualizing genomic characteristics across an RNA-Seq based reference landscape of normal and neoplastic brain.
- Author
-
Arora S, Szulzewsky F, Jensen M, Nuechterlein N, Pattwell SS, and Holland EC
- Subjects
- Adult, Child, Humans, RNA-Seq, Brain, Genomics, Brain Neoplasms genetics, Glioma
- Abstract
In order to better understand the relationship between normal and neoplastic brain, we combined five publicly available large-scale datasets, correcting for batch effects and applying Uniform Manifold Approximation and Projection (UMAP) to RNA-Seq data. We assembled a reference Brain-UMAP including 702 adult gliomas, 802 pediatric tumors and 1409 healthy normal brain samples, which can be utilized to investigate the wealth of information obtained from combining several publicly available datasets to study a single organ site. Normal brain regions and tumor types create distinct clusters and because the landscape is generated by RNA-Seq, comparative gene expression profiles and gene ontology patterns are readily evident. To our knowledge, this is the first meta-analysis that allows for comparison of gene expression and pathways of interest across adult gliomas, pediatric brain tumors, and normal brain regions. We provide access to this resource via the open source, interactive online tool Oncoscape, where the scientific community can readily visualize clinical metadata, gene expression patterns, gene fusions, mutations, and copy number patterns for individual genes and pathway over this reference landscape., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
16. Nuclear condensates of YAP fusion proteins alter transcription to drive ependymoma tumourigenesis.
- Author
-
Hu X, Wu X, Berry K, Zhao C, Xin D, Ogurek S, Liu X, Zhang L, Luo Z, Sakabe M, Trubicka J, Łastowska M, Szulzewsky F, Holland EC, Lee L, Hu M, Xin M, and Lu QR
- Subjects
- Humans, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Carcinogenesis genetics, Cell Cycle Proteins metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Nuclear Proteins metabolism, YAP-Signaling Proteins, Cell Nucleus metabolism, Transcription, Genetic, Ependymoma genetics, Ependymoma metabolism, Ependymoma pathology, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Nuclear localization of HIPPO-YAP fusion proteins has been implicated in supratentorial ependymoma development. Here, unexpectedly, we find that liquid-liquid phase separation, rather than nuclear localization, of recurrent patient-derived YAP fusions, YAP-MAMLD1 and C11ORF95-YAP, underlies ependymoma tumourigenesis from neural progenitor cells. Mutagenesis and chimaera assays demonstrate that an intrinsically disordered region promotes oligomerization of the YAP fusions into nuclear, puncta-like, membrane-less condensates. Oligomerization and nuclear condensates induced by YAP fusion with a coiled-coil domain of transcriptional activator GCN4 also promote ependymoma formation. YAP-MAMLD1 concentrates transcription factors and co-activators, including BRD4, MED1 and TEAD, in condensates while excluding transcriptional repressive PRC2, and induces long-range enhancer-promoter interactions that promote transcription and oncogenic programmes. Blocking condensate-mediated transcriptional co-activator activity inhibits tumourigenesis, indicating a critical role of liquid phase separation for YAP fusion oncogenic activity in ependymoma. YAP fusions containing the intrinsically disordered region features are common in human tumours, suggesting that nuclear condensates could be targeted to treat YAP-fusion-induced cancers., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
17. An RNA seq-based reference landscape of human normal and neoplastic brain.
- Author
-
Arora S, Szulzewsky F, Jensen M, Nuechterlein N, Pattwell SS, and Holland EC
- Abstract
In order to better understand the relationship between normal and neoplastic brain, we combined five publicly available large-scale datasets, correcting for batch effects and applying Uniform Manifold Approximation and Projection (UMAP) to RNA-seq data. We assembled a reference Brain-UMAP including 702 adult gliomas, 802 pediatric tumors and 1409 healthy normal brain samples, which can be utilized to investigate the wealth of information obtained from combining several publicly available datasets to study a single organ site. Normal brain regions and tumor types create distinct clusters and because the landscape is generated by RNA seq, comparative gene expression profiles and gene ontology patterns are readily evident. To our knowledge, this is the first meta-analysis that allows for comparison of gene expression and pathways of interest across adult gliomas, pediatric brain tumors, and normal brain regions. We provide access to this resource via the open source, interactive online tool Oncoscape, where the scientific community can readily visualize clinical metadata, gene expression patterns, gene fusions, mutations, and copy number patterns for individual genes and pathway over this reference landscape.
- Published
- 2023
- Full Text
- View/download PDF
18. Oncogenic role of a developmentally regulated NTRK2 splice variant.
- Author
-
Pattwell SS, Arora S, Nuechterlein N, Zager M, Loeb KR, Cimino PJ, Holland NC, Reche-Ley N, Bolouri H, Almiron Bonnin DA, Szulzewsky F, Phadnis VV, Ozawa T, Wagner MJ, Haffner MC, Cao J, Shendure J, and Holland EC
- Abstract
Temporally regulated alternative splicing choices are vital for proper development, yet the wrong splice choice may be detrimental. Here, we highlight a previously unidentified role for the neurotrophin receptor splice variant TrkB.T1 in neurodevelopment, embryogenesis, transformation, and oncogenesis across multiple tumor types in humans and mice. TrkB.T1 is the predominant NTRK2 isoform across embryonic organogenesis, and forced overexpression of this embryonic pattern causes multiple solid and nonsolid tumors in mice in the context of tumor suppressor loss. TrkB.T1 also emerges as the predominant NTRK isoform expressed in a wide range of adult and pediatric tumors, including those harboring tropomyosin receptor kinase fusions. Affinity purification-mass spectrometry proteomic analysis reveals distinct interactors with known developmental and oncogenic signaling pathways such as Wnt, transforming growth factor-β, Sonic Hedgehog, and Ras. From alterations in splicing factors to changes in gene expression, the discovery of isoform specific oncogenes with embryonic ancestry has the potential to shape the way we think about developmental systems and oncology.
- Published
- 2022
- Full Text
- View/download PDF
19. Both YAP1-MAML2 and constitutively active YAP1 drive the formation of tumors that resemble NF2 mutant meningiomas in mice.
- Author
-
Szulzewsky F, Arora S, Arakaki AKS, Sievers P, Almiron Bonnin DA, Paddison PJ, Sahm F, Cimino PJ, Gujral TS, and Holland EC
- Abstract
YAP1 is a transcriptional coactivator regulated by the Hippo signaling pathway, including NF2. Meningiomas are the most common primary brain tumors; a large percentage exhibit heterozygous loss of chromosome 22 (harboring the NF2 gene) and functional inactivation of the remaining NF2 copy, implicating oncogenic YAP activity in these tumors. Recently, fusions between YAP1 and MAML2 have been identified in a subset of pediatric NF2 wild-type meningiomas. Here, we show that human YAP1-MAML2 -positive meningiomas resemble NF2 mutant meningiomas by global and YAP-related gene expression signatures. We then show that expression of YAP1-MAML2 in mice induces tumors that resemble human YAP1 fusion-positive and NF2 mutant meningiomas by gene expression. We demonstrate that YAP1-MAML2 primarily functions by exerting TEAD-dependent YAP activity that is resistant to Hippo signaling. Treatment with YAP-TEAD inhibitors is sufficient to inhibit the viability of YAP1-MAML2 -driven mouse tumors ex vivo. Finally, we show that expression of constitutively active YAP1 (S127/397A-YAP1) is sufficient to induce similar tumors, suggesting that the YAP component of the gene fusion is the critical driver of these tumors. In summary, our results implicate YAP1-MAML2 as a causal oncogenic driver and highlight TEAD-dependent YAP activity as an oncogenic driver in YAP1-MAML2 fusion meningioma as well as NF2 mutant meningioma in general., (© 2022 Szulzewsky et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
20. Utilizing preclinical models to develop targeted therapies for rare central nervous system cancers.
- Author
-
Arakaki AKS, Szulzewsky F, Gilbert MR, Gujral TS, and Holland EC
- Subjects
- Central Nervous System, Humans, Central Nervous System Neoplasms drug therapy, Central Nervous System Neoplasms genetics, Ependymoma, Glioma, Rhabdoid Tumor
- Abstract
Patients with rare central nervous system (CNS) tumors typically have a poor prognosis and limited therapeutic options. Historically, these cancers have been difficult to study due to small number of patients. Recent technological advances have identified molecular drivers of some of these rare cancers which we can now use to generate representative preclinical models of these diseases. In this review, we outline the advantages and disadvantages of different models, emphasizing the utility of various in vitro and ex vivo models for target discovery and mechanistic inquiry and multiple in vivo models for therapeutic validation. We also highlight recent literature on preclinical model generation and screening approaches for ependymomas, histone mutated high-grade gliomas, and atypical teratoid rhabdoid tumors, all of which are rare CNS cancers that have recently established genetic or epigenetic drivers. These preclinical models are critical to advancing targeted therapeutics for these rare CNS cancers that currently rely on conventional treatments., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2021
- Full Text
- View/download PDF
21. Leveraging the replication-competent avian-like sarcoma virus/tumor virus receptor-A system for modeling human gliomas.
- Author
-
Kanvinde PP, Malla AP, Connolly NP, Szulzewsky F, Anastasiadis P, Ames HM, Kim AJ, Winkles JA, Holland EC, and Woodworth GF
- Subjects
- Animals, Avian Sarcoma Viruses genetics, Humans, Oncogenic Viruses, Receptors, Virus, Tumor Microenvironment, Brain Neoplasms pathology, Disease Models, Animal, Glioma pathology, Sarcoma
- Abstract
Gliomas are the most common primary intrinsic brain tumors occurring in adults. Of all malignant gliomas, glioblastoma (GBM) is considered the deadliest tumor type due to diffuse brain invasion, immune evasion, cellular, and molecular heterogeneity, and resistance to treatments resulting in high rates of recurrence. An extensive understanding of the genomic and microenvironmental landscape of gliomas gathered over the past decade has renewed interest in pursuing novel therapeutics, including immune checkpoint inhibitors, glioma-associated macrophage/microglia (GAMs) modulators, and others. In light of this, predictive animal models that closely recreate the conditions and findings found in human gliomas will serve an increasingly important role in identifying new, effective therapeutic strategies. Although numerous syngeneic, xenograft, and transgenic rodent models have been developed, few include the full complement of pathobiological features found in human tumors, and therefore few accurately predict bench-to-bedside success. This review provides an update on how genetically engineered rodent models based on the replication-competent avian-like sarcoma (RCAS) virus/tumor virus receptor-A (tv-a) system have been used to recapitulate key elements of human gliomas in an immunologically intact host microenvironment and highlights new approaches using this model system as a predictive tool for advancing translational glioma research., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
22. YAP1 and its fusion proteins in cancer initiation, progression and therapeutic resistance.
- Author
-
Szulzewsky F, Holland EC, and Vasioukhin V
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic genetics, Humans, Neoplasms genetics, Oncogene Proteins, Fusion genetics, Oncogenes genetics, Phosphoproteins genetics, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Signal Transduction genetics, Transcription Factors genetics, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing physiology, Oncogene Proteins, Fusion metabolism, Transcription Factors metabolism, Transcription Factors physiology
- Abstract
YAP1 is a transcriptional co-activator whose activity is controlled by the Hippo signaling pathway. In addition to important functions in normal tissue homeostasis and regeneration, YAP1 has also prominent functions in cancer initiation, aggressiveness, metastasis, and therapy resistance. In this review we are discussing the molecular functions of YAP1 and its roles in cancer, with a focus on the different mechanisms of de-regulation of YAP1 activity in human cancers, including inactivation of upstream Hippo pathway tumor suppressors, regulation by intersecting pathways, miRNAs, and viral oncogenes. We are also discussing new findings on the function and biology of the recently identified family of YAP1 gene fusions, that constitute a new type of activating mutation of YAP1 and that are the likely oncogenic drivers in several subtypes of human cancers. Lastly, we also discuss different strategies of therapeutic inhibition of YAP1 functions., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Publisher Correction to: C11orf95-RELA fusion drives aberrant gene expression through the unique epigenetic regulation for ependymoma formation.
- Author
-
Ozawa T, Kaneko S, Szulzewsky F, Qiao Z, Takadera M, Narita Y, Kondo T, Holland EC, Hamamoto R, and Ichimura K
- Published
- 2021
- Full Text
- View/download PDF
24. C11orf95-RELA fusion drives aberrant gene expression through the unique epigenetic regulation for ependymoma formation.
- Author
-
Ozawa T, Kaneko S, Szulzewsky F, Qiao Z, Takadera M, Narita Y, Kondo T, Holland EC, Hamamoto R, and Ichimura K
- Subjects
- Animals, Brain Neoplasms genetics, Gene Expression, Gene Expression Regulation, Genetic Techniques, HEK293 Cells, Humans, Mice, NF-kappa B metabolism, Supratentorial Neoplasms genetics, DNA-Binding Proteins genetics, Ependymoma genetics, Epigenesis, Genetic, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Proteins genetics, Transcription Factor RelA genetics
- Abstract
Recurrent C11orf95-RELA fusions (RELA
FUS ) are the hallmark of supratentorial ependymomas. The presence of RELA as the fusion partner indicates a close association of aberrant NF-κB activity with tumorigenesis. However, the oncogenic role of the C11orf95 has not been determined. Here, we performed ChIP-seq analyses to explore genomic regions bound by RELAFUS and H3K27ac proteins in human 293T and mouse ependymoma cells. We then utilized published RNA-Seq data from human and mouse RELAFUS tumors and identified target genes that were directly regulated by RELAFUS in these tumors. Subsequent transcription factor motif analyses of RELAFUS target genes detected a unique GC-rich motif recognized by the C11orf95 moiety, that is present in approximately half of RELAFUS target genes. Luciferase assays confirmed that a promoter carrying this motif is sufficient to drive RELAFUS -dependent gene expression. Further, the RELAFUS target genes were found to be overlapped with Rela target genes primarily via non-canonical NF-κB binding sites. Using a series of truncation and substitution mutants of RELAFUS , we also show that the activation domain in the RELAFUS moiety is necessary for the regulation of gene expression of these RELAFUS target genes. Lastly, we performed an anti-cancer drug screening with mouse ependymoma cells and identified potential anti-ependymoma drugs that are related to the oncogenic mechanism of RELAFUS . These findings suggested that RELAFUS might induce ependymoma formation through oncogenic pathways orchestrated by both C11orf95 and RELA target genes. Thus, our study unveils a complex gene function of RELAFUS as an oncogenic transcription factor in RELAFUS positive ependymomas.- Published
- 2021
- Full Text
- View/download PDF
25. Platelet-derived growth factor beta is a potent inflammatory driver in paediatric high-grade glioma.
- Author
-
Ross JL, Chen Z, Herting CJ, Grabovska Y, Szulzewsky F, Puigdelloses M, Monterroza L, Switchenko J, Wadhwani NR, Cimino PJ, Mackay A, Jones C, Read RD, MacDonald TJ, Schniederjan M, Becher OJ, and Hambardzumyan D
- Subjects
- Adolescent, Adult, Animals, Brain Neoplasms genetics, Cells, Cultured, Chemokines genetics, Child, Child, Preschool, Encephalitis genetics, Female, Glioma, Humans, Infant, Infant, Newborn, Male, Mice, Inbred C57BL, Transcriptome, Young Adult, Mice, Brain Neoplasms immunology, Encephalitis immunology, Proto-Oncogene Proteins c-sis immunology, Tumor-Associated Macrophages immunology
- Abstract
Paediatric high-grade gliomas (HGGs) account for the most brain tumour-related deaths in children and have a median survival of 12-15 months. One promising avenue of research is the development of novel therapies targeting the properties of non-neoplastic cell-types within the tumour such as tumour associated macrophages (TAMs). TAMs are immunosuppressive and promote tumour malignancy in adult HGG; however, in paediatric medulloblastoma, TAMs exhibit anti-tumour properties. Much is known about TAMs in adult HGG, yet little is known about them in the paediatric setting. This raises the question of whether paediatric HGGs possess a distinct constituency of TAMs because of their unique genetic landscapes. Using human paediatric HGG tissue samples and murine models of paediatric HGG, we demonstrate diffuse midline gliomas possess a greater inflammatory gene expression profile compared to hemispheric paediatric HGGs. We also show despite possessing sparse T-cell infiltration, human paediatric HGGs possess high infiltration of IBA1+ TAMs. CD31, PDGFRβ, and PDGFB all strongly correlate with IBA1+ TAM infiltration. To investigate the TAM population, we used the RCAS/tv-a system to recapitulate paediatric HGG in newborn immunocompetent mice. Tumours are induced in Nestin-positive brain cells by PDGFA or PDGFB overexpression with Cdkn2a or Tp53 co-mutations. Tumours driven by PDGFB have a significantly lower median survival compared to PDGFA-driven tumours and have increased TAM infiltration. NanoString and quantitative PCR analysis indicates PDGFB-driven tumours have a highly inflammatory microenvironment characterized by high chemokine expression. In vitro bone marrow-derived monocyte and microglial cultures demonstrate bone marrow-derived monocytes are most responsible for the production of inflammatory signals in the tumour microenvironment in response to PDGFB stimulation. Lastly, using knockout mice deficient for individual chemokines, we demonstrate the feasibility of reducing TAM infiltration and prolonging survival in both PDGFA and PDGFB-driven tumours. We identify CCL3 as a potential key chemokine in these processes in both humans and mice. Together, these studies provide evidence for the potent inflammatory effects PDGFB has in paediatric HGGs., (© The Author(s) (2020). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
26. Multimodal single-cell analysis reveals distinct radioresistant stem-like and progenitor cell populations in murine glioma.
- Author
-
Alexander J, LaPlant QC, Pattwell SS, Szulzewsky F, Cimino PJ, Caruso FP, Pugliese P, Chen Z, Chardon F, Hill AJ, Spurrell C, Ahrendsen D, Pietras A, Starita LM, Hambardzumyan D, Iavarone A, Shendure J, and Holland EC
- Subjects
- Animals, Mice, Neoplastic Stem Cells, Single-Cell Analysis, Tumor Microenvironment, Brain Neoplasms radiotherapy, Glioma, Stem Cells
- Abstract
Radiation therapy is part of the standard of care for gliomas and kills a subset of tumor cells, while also altering the tumor microenvironment. Tumor cells with stem-like properties preferentially survive radiation and give rise to glioma recurrence. Various techniques for enriching and quantifying cells with stem-like properties have been used, including the fluorescence activated cell sorting (FACS)-based side population (SP) assay, which is a functional assay that enriches for stem-like tumor cells. In these analyses, mouse models of glioma have been used to understand the biology of this disease and therapeutic responses, including the radiation response. We present combined SP analysis and single-cell RNA sequencing of genetically-engineered mouse models of glioma to show a time course of cellular response to radiation. We identify and characterize two distinct tumor cell populations that are inherently radioresistant and also distinct effects of radiation on immune cell populations within the tumor microenvironment., (© 2020 The Authors. Glia published by Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
27. Phenotypic characterization with somatic genome editing and gene transfer reveals the diverse oncogenicity of ependymoma fusion genes.
- Author
-
Takadera M, Satomi K, Szulzewsky F, Cimino PJ, Holland EC, Yamamoto T, Ichimura K, and Ozawa T
- Subjects
- Animals, CRISPR-Cas Systems, Cells, Cultured, Chromothripsis, DNA Copy Number Variations, Gene Editing, Gene Transfer Techniques, Mice, Phenotype, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing genetics, Brain Neoplasms genetics, Carcinogenesis genetics, DNA-Binding Proteins genetics, Ependymoma genetics, Oncogene Fusion genetics, Transcription Factor RelA genetics, Transcription Factors genetics
- Abstract
Recurrent RELA and YAP1 fusions are intimately associated with tumorigenesis in supratentorial ependymomas. Chromothripsis and focal copy number alterations involving 11q are hallmarks of these tumors. However, it is unknown whether the chromosomal alterations are a direct causal event resulting in fusion transcripts. In addition, the biological significance of the RELA fusion variants and YAP1 fusions is not yet fully characterized. In this study, we generated gene rearrangements on 11q with the CRISPR/Cas9 system and investigated the formation of oncogenic ependymoma fusion genes. Further, we examined the oncogenic potential of RELA fusion variants and YAP1 fusions in a lentiviral gene transfer model. We observed that endogenous RELA fusion events were successfully induced by CRISPR/Cas9-mediated genome rearrangement in cultured cells. In vivo genome editing in mouse brain resulted in the development of ependymoma-like brain tumors that harbored the Rela fusion gene. All RELA fusion variants tested, except a variant lacking the Rel homology domain, were able to induce tumor formation, albeit with different efficacy. Furthermore, expression of YAP1-FAM118B and YAP1-MAMLD1 fusions induced the formation of spindle-cell-like tumors at varying efficacy. Our results indicate that chromosomal rearrangements involving the Rela locus are the causal event for the formation of Rela fusion-driven ependymomas in mice. Furthermore, the type of RELA. fusion might affect the aggressiveness of tumors and that the Rel homology domain is essential for the oncogenic functions of RELA. fusions. The YAP1 fusion genes are also oncogenic when expressed in mice.
- Published
- 2020
- Full Text
- View/download PDF
28. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression.
- Author
-
De Boeck A, Ahn BY, D'Mello C, Lun X, Menon SV, Alshehri MM, Szulzewsky F, Shen Y, Khan L, Dang NH, Reichardt E, Goring KA, King J, Grisdale CJ, Grinshtein N, Hambardzumyan D, Reilly KM, Blough MD, Cairncross JG, Yong VW, Marra MA, Jones SJM, Kaplan DR, McCoy KD, Holland EC, Bose P, Chan JA, Robbins SM, and Senger DL
- Subjects
- Animals, Brain Neoplasms mortality, Carcinogenesis, Cell Nucleus metabolism, Cytokines metabolism, Glioblastoma metabolism, Glioblastoma mortality, Glioblastoma pathology, Glioma mortality, Humans, Inflammation, Killer Cells, Natural metabolism, Killer Cells, Natural pathology, Macrophages metabolism, Macrophages pathology, Mice, Mice, SCID, Microglia, Survival Analysis, T-Lymphocytes metabolism, T-Lymphocytes pathology, Tumor Microenvironment immunology, Brain Neoplasms metabolism, Brain Neoplasms pathology, Glioma metabolism, Glioma pathology, Interleukin-33 metabolism
- Abstract
Despite a deeper molecular understanding, human glioblastoma remains one of the most treatment refractory and fatal cancers. It is known that the presence of macrophages and microglia impact glioblastoma tumorigenesis and prevent durable response. Herein we identify the dual function cytokine IL-33 as an orchestrator of the glioblastoma microenvironment that contributes to tumorigenesis. We find that IL-33 expression in a large subset of human glioma specimens and murine models correlates with increased tumor-associated macrophages/monocytes/microglia. In addition, nuclear and secreted functions of IL-33 regulate chemokines that collectively recruit and activate circulating and resident innate immune cells creating a pro-tumorigenic environment. Conversely, loss of nuclear IL-33 cripples recruitment, dramatically suppresses glioma growth, and increases survival. Our data supports the paradigm that recruitment and activation of immune cells, when instructed appropriately, offer a therapeutic strategy that switches the focus from the cancer cell alone to one that includes the normal host environment.
- Published
- 2020
- Full Text
- View/download PDF
29. Genetic driver mutations introduced in identical cell-of-origin in murine glioblastoma reveal distinct immune landscapes but similar response to checkpoint blockade.
- Author
-
Chen Z, Herting CJ, Ross JL, Gabanic B, Puigdelloses Vallcorba M, Szulzewsky F, Wojciechowicz ML, Cimino PJ, Ezhilarasan R, Sulman EP, Ying M, Ma'ayan A, Read RD, and Hambardzumyan D
- Subjects
- Animals, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Female, Glioblastoma drug therapy, Glioblastoma pathology, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Tumor Cells, Cultured, Brain Neoplasms genetics, Brain Neoplasms immunology, Glioblastoma genetics, Glioblastoma immunology, Immune Checkpoint Inhibitors therapeutic use, Mutation physiology
- Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor. In addition to being genetically heterogeneous, GBMs are also immunologically heterogeneous. However, whether the differences in immune microenvironment are driven by genetic driver mutation is unexplored. By leveraging the versatile RCAS/tv-a somatic gene transfer system, we establish a mouse model for Classical GBM by introducing EGFRvIII expression in Nestin-positive neural stem/progenitor cells in adult mice. Along with our previously published Nf1-silenced and PDGFB-overexpressing models, we investigate the immune microenvironments of the three models of human GBM subtypes by unbiased multiplex profiling. We demonstrate that both the quantity and composition of the microenvironmental myeloid cells are dictated by the genetic driver mutations, closely mimicking what was observed in human GBM subtypes. These myeloid cells express high levels of the immune checkpoint protein PD-L1; however, PD-L1 targeted therapies alone or in combination with irradiation are unable to increase the survival time of tumor-bearing mice regardless of the driver mutations, reflecting the outcomes of recent human trials. Together, these results highlight the critical utility of immunocompetent mouse models for preclinical studies of GBM, making these models indispensable tools for understanding the resistance mechanisms of immune checkpoint blockade in GBM and immune cell-targeting drug discovery., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
30. Mathematical modeling of PDGF-driven glioma reveals the dynamics of immune cells infiltrating into tumors.
- Author
-
Niu B, Zeng X, Phan TA, Szulzewsky F, Holte S, Holland EC, and Tian JP
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms pathology, Glioma genetics, Glioma metabolism, Glioma pathology, Humans, Lymphocytes, Tumor-Infiltrating pathology, Mice, Brain Neoplasms immunology, Glioma immunology, Isocitrate Dehydrogenase genetics, Lymphocytes, Tumor-Infiltrating immunology, Models, Theoretical, Mutation, Platelet-Derived Growth Factor metabolism
- Abstract
Background: Tumor-infiltrated immune cells compose a significant component of many cancers. They have been observed to have contradictory impacts on tumors. Although the primary reasons for these observations remain elusive, it is important to understand how immune cells infiltrating into tumors is regulated. Recently our group conducted a series of experimental studies, which showed that muIDH1 gliomas have a significant global reduction of immune cells and suggested that the longer survival time of mice with CIMP gliomas may be due to the IDH mutation and its effect on reducing of the tumor-infiltrated immune cells. However, to comprehend how IDH1 mutants regulate infiltration of immune cells into gliomas and how they affect the aggressiveness of gliomas, it is necessary to integrate our experimental data into a dynamical system to acquire a much deeper understanding of subtle regulation of immune cell infiltration., Methods: The method is integration of mathematical modeling and experiments. According to mass conservation laws and assumption that immune cells migrate into the tumor site along a chemotactic gradient field, a mathematical model is formulated. Parameters are estimated from our experiments. Numerical methods are developed to solve the problem. Numerical predictions are compared with experimental results., Results: Our analysis shows that the net rate of increase of immune cells infiltrated into the tumor is approximately proportional to the 4/5 power of the chemoattractant production rate, and it is an increasing function of time while the percentage of immune cells infiltrated into the tumor is a decreasing function of time. Our model predicts that wtIDH1 mice will survive longer if the immune cells are blocked by reducing chemotactic coefficient. For more aggressive gliomas, our model shows that there is little difference in their survivals between wtIDH1 and muIDH1 tumors, and the percentage of immune cells infiltrated into the tumor is much lower. These predictions are verified by our experimental results. In addition, wtIDH1 and muIDH1 can be quantitatively distinguished by their chemoattractant production rates, and the chemotactic coefficient determines possibilities of immune cells migration along chemoattractant gradient fields., Conclusions: The chemoattractant gradient field produced by tumor cells may facilitate immune cells migration to the tumor cite. The chemoattractant production rate may be utilized to classify wtIDH1 and muIDH1 tumors. The dynamics of immune cells infiltrating into tumors is largely determined by tumor cell chemoattractant production rate and chemotactic coefficient., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
31. Comparison of tumor-associated YAP1 fusions identifies a recurrent set of functions critical for oncogenesis.
- Author
-
Szulzewsky F, Arora S, Hoellerbauer P, King C, Nathan E, Chan M, Cimino PJ, Ozawa T, Kawauchi D, Pajtler KW, Gilbertson RJ, Paddison PJ, Vasioukhin V, Gujral TS, and Holland EC
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Cells, Cultured, Gene Expression Regulation, Humans, Mice, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Nuclear Localization Signals, Nucleotide Motifs, Oncogene Proteins, Fusion antagonists & inhibitors, Oncogene Proteins, Fusion chemistry, Proteasome Endopeptidase Complex metabolism, Signal Transduction, Transcription Factors metabolism, Transcription, Genetic, Adaptor Proteins, Signal Transducing metabolism, Carcinogenesis genetics, Oncogene Proteins, Fusion metabolism
- Abstract
YAP1 is a transcriptional coactivator and the principal effector of the Hippo signaling pathway, which is causally implicated in human cancer. Several YAP1 gene fusions have been identified in various human cancers and identifying the essential components of this family of gene fusions has significant therapeutic value. Here, we show that the YAP1 gene fusions YAP1-MAMLD1 , YAP1-FAM118B , YAP1-TFE3 , and YAP1-SS18 are oncogenic in mice. Using reporter assays, RNA-seq, ChIP-seq, and loss-of-function mutations, we can show that all of these YAP1 fusion proteins exert TEAD-dependent YAP activity, while some also exert activity of the C'-terminal fusion partner. The YAP activity of the different YAP1 fusions is resistant to negative Hippo pathway regulation due to constitutive nuclear localization and resistance to degradation of the YAP1 fusion proteins. Genetic disruption of the TEAD-binding domain of these oncogenic YAP1 fusions is sufficient to inhibit tumor formation in vivo, while pharmacological inhibition of the YAP1-TEAD interaction inhibits the growth of YAP1 fusion-expressing cell lines in vitro. These results highlight TEAD-dependent YAP activity found in these gene fusions as critical for oncogenesis and implicate these YAP functions as potential therapeutic targets in YAP1 fusion-positive tumors., (© 2020 Szulzewsky et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2020
- Full Text
- View/download PDF
32. Fusing the Genetic Landscape of Infantile High-Grade Gliomas.
- Author
-
Szulzewsky F and Cimino PJ
- Subjects
- Gene Fusion, Humans, Infant, Proto-Oncogene Proteins genetics, Receptor Protein-Tyrosine Kinases genetics, Glioma genetics, Protein-Tyrosine Kinases genetics
- Abstract
In this issue of Cancer Discovery , Clarke and colleagues define the genetic landscape of infantile cerebral high-grade gliomas, which frequently contain alterations in the MAPK pathway, as well as recurrent gene fusions in receptor tyrosine kinases ( ALK, ROS1, MET ) and neurotrophic receptor kinases ( NTRK1-3 ). Combining their multi-omic profiling data with functional preclinical and clinical studies, this large multi-institutional study provides strong rationale for future classification and molecular subtype-specific therapeutic management of infantile high-grade glioma. See related article by Clarke et al., p. 942 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
33. A kinase-deficient NTRK2 splice variant predominates in glioma and amplifies several oncogenic signaling pathways.
- Author
-
Pattwell SS, Arora S, Cimino PJ, Ozawa T, Szulzewsky F, Hoellerbauer P, Bonifert T, Hoffstrom BG, Boiani NE, Bolouri H, Correnti CE, Oldrini B, Silber JR, Squatrito M, Paddison PJ, and Holland EC
- Subjects
- Animals, Brain metabolism, Brain pathology, Brain Neoplasms metabolism, Brain Neoplasms pathology, Carcinogenesis genetics, Cells, Cultured, Gene Expression Profiling, Gene Ontology, Glioma metabolism, Glioma pathology, High-Throughput Nucleotide Sequencing, Humans, Membrane Glycoproteins metabolism, Mice, NIH 3T3 Cells, Neural Stem Cells metabolism, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Phosphatidylinositol 3-Kinases metabolism, RNA Isoforms metabolism, Receptor, trkB metabolism, Signal Transduction genetics, Brain Neoplasms genetics, Glioma genetics, Membrane Glycoproteins genetics, Oncogenes genetics, RNA Isoforms genetics, RNA Splicing, Receptor, trkB genetics
- Abstract
Independent scientific achievements have led to the discovery of aberrant splicing patterns in oncogenesis, while more recent advances have uncovered novel gene fusions involving neurotrophic tyrosine receptor kinases (NTRKs) in gliomas. The exploration of NTRK splice variants in normal and neoplastic brain provides an intersection of these two rapidly evolving fields. Tropomyosin receptor kinase B (TrkB), encoded NTRK2, is known for critical roles in neuronal survival, differentiation, molecular properties associated with memory, and exhibits intricate splicing patterns and post-translational modifications. Here, we show a role for a truncated NTRK2 splice variant, TrkB.T1, in human glioma. TrkB.T1 enhances PDGF-driven gliomas in vivo, augments PDGF-induced Akt and STAT3 signaling in vitro, while next generation sequencing broadly implicates TrkB.T1 in the PI3K signaling cascades in a ligand-independent fashion. These TrkB.T1 findings highlight the importance of expanding upon whole gene and gene fusion analyses to include splice variants in basic and translational neuro-oncology research.
- Published
- 2020
- Full Text
- View/download PDF
34. Anti-PD-L1 antibody direct activation of macrophages contributes to a radiation-induced abscopal response in glioblastoma.
- Author
-
Ene CI, Kreuser SA, Jung M, Zhang H, Arora S, White Moyes K, Szulzewsky F, Barber J, Cimino PJ, Wirsching HG, Patel A, Kong P, Woodiwiss TR, Durfy SJ, Houghton AM, Pierce RH, Parney IF, Crane CA, and Holland EC
- Subjects
- Animals, B7-H1 Antigen, Immunotherapy, Macrophages, Glioblastoma radiotherapy, Glioma drug therapy, Glioma radiotherapy
- Abstract
Background: Most glioblastomas recur near prior radiation treatment sites. Future clinical success will require achieving and optimizing an "abscopal effect," whereby unirradiated neoplastic cells outside treatment sites are recognized and attacked by the immune system. Radiation combined with anti-programmed cell death ligand 1 (PD-L1) demonstrated modest efficacy in phase II human glioblastoma clinical trials, but the mechanism and relevance of the abscopal effect during this response remain unknown., Methods: We modified an immune-competent, genetically driven mouse glioma model (forced platelet derived growth factor [PDGF] expression + phosphatase and tensin homolog loss) where a portion of the tumor burden is irradiated (PDGF) and another unirradiated luciferase-expressing tumor (PDGF + luciferase) is used as a readout of the abscopal effect following systemic anti-PD-L1 immunotherapy. We assessed relevance of tumor neoepitope during the abscopal response by inducing expression of epidermal growth factor receptor variant III (EGFRvIII) (PDGF + EGFRvIII). Statistical tests were two-sided., Results: Following radiation of one lesion, anti-PD-L1 immunotherapy enhanced the abscopal response to the unirradiated lesion. In PDGF-driven gliomas without tumor neoepitope (PDGF + luciferase, n = 8), the abscopal response occurred via anti-PD-L1 driven, extracellular signal-regulated kinase-mediated, bone marrow-derived macrophage phagocytosis of adjacent unirradiated tumor cells, with modest survival implications (median survival 41 days vs radiation alone 37.5 days, P = 0.03). In PDGF-driven gliomas with tumor neoepitope (PDGF + EGFRvIII, n = 8), anti-PD-L1 enhanced abscopal response was associated with macrophage and T-cell infiltration and increased survival benefit (median survival 36 days vs radiation alone 28 days, P = 0.001)., Conclusion: Our results indicate that anti-PD-L1 immunotherapy enhances a radiation- induced abscopal response via canonical T-cell activation and direct macrophage activation in glioblastoma., (© The Author(s) 2019. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
35. Cooperation of oncolytic virotherapy with VEGF-neutralizing antibody treatment in IDH wildtype glioblastoma depends on MMP9.
- Author
-
Wirsching HG, Arora S, Zhang H, Szulzewsky F, Cimino PJ, Quéva C, Houghton AM, Glorioso JC, Weller M, and Holland EC
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms pathology, Combined Modality Therapy, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma pathology, Humans, Isocitrate Dehydrogenase genetics, Mice, Survival Rate, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A immunology, Xenograft Model Antitumor Assays, Antibodies, Neutralizing pharmacology, Brain Neoplasms therapy, Glioblastoma therapy, Matrix Metalloproteinase 9 metabolism, Mutation, Oncolytic Virotherapy mortality, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Published
- 2019
- Full Text
- View/download PDF
36. Tumour-associated macrophage-derived interleukin-1 mediates glioblastoma-associated cerebral oedema.
- Author
-
Herting CJ, Chen Z, Maximov V, Duffy A, Szulzewsky F, Shayakhmetov DM, and Hambardzumyan D
- Subjects
- Animals, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Bone Marrow Cells pathology, Brain Edema etiology, Brain Edema pathology, Brain Neoplasms complications, Brain Neoplasms pathology, Dexamethasone pharmacology, Glioblastoma complications, Glioblastoma pathology, Glucocorticoids pharmacology, Macrophages drug effects, Macrophages pathology, Mice, Microglia drug effects, Microglia metabolism, Signal Transduction drug effects, Brain Edema metabolism, Brain Neoplasms metabolism, Glioblastoma metabolism, Interleukin-1 metabolism, Macrophages metabolism
- Abstract
Glioblastoma is the most common and uncompromising primary brain tumour and is characterized by a dismal prognosis despite aggressive treatment regimens. At the cellular level, these tumours are composed of a mixture of neoplastic cells and non-neoplastic cells, including tumour-associated macrophages and endothelial cells. Cerebral oedema is a near-universal occurrence in patients afflicted with glioblastoma and it is almost exclusively managed with the corticosteroid dexamethasone despite significant drawbacks associated with its use. Here, we demonstrate that dexamethasone blocks interleukin-1 production in both bone marrow-derived and brain resident macrophage populations following stimulation with lipopolysaccharide and interferon gamma. Additionally, dexamethasone is shown to inhibit downstream effectors of interleukin-1 signalling in both macrophage populations. Co-culture of bone marrow-derived macrophages with organotypic tumour slices results in an upregulation of interleukin-1 cytokines, an effect that is absent in co-cultured microglia. Genetic ablation of interleukin-1 ligands or receptor in mice bearing RCAS/tv-a-induced platelet-derived growth factor B-overexpressing glioblastoma results in reduced oedema and partial restoration of the integrity of the blood-brain barrier, respectively; similar to results obtained with vascular endothelial growth factor neutralization. We establish that tumours from dexamethasone-treated mice exhibit reduced infiltration of cells of the myeloid and lymphoid compartments, an effect that should be considered during clinical trials for immunotherapy in glioblastoma patients. Additionally, we emphasize that caution should be used when immune profiling and single-cell RNA sequencing data are interpreted from fresh glioblastoma patient samples, as nearly all patients receive dexamethasone after diagnosis. Collectively, this evidence suggests that interleukin-1 signalling inhibition and dexamethasone treatment share therapeutic efficacies and establishes interleukin-1 signalling as an attractive and specific therapeutic target for the management of glioblastoma-associated cerebral oedema., (© The Author(s) (2019). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
37. Human Mesenchymal glioblastomas are characterized by an increased immune cell presence compared to Proneural and Classical tumors.
- Author
-
Kaffes I, Szulzewsky F, Chen Z, Herting CJ, Gabanic B, Velázquez Vega JE, Shelton J, Switchenko JM, Ross JL, McSwain LF, Huse JT, Westermark B, Nelander S, Forsberg-Nilsson K, Uhrbom L, Maturi NP, Cimino PJ, Holland EC, Kettenmann H, Brennan CW, Brat DJ, and Hambardzumyan D
- Abstract
Glioblastoma (GBM) is the most aggressive malignant primary brain tumor in adults, with a median survival of 14.6 months. Recent efforts have focused on identifying clinically relevant subgroups to improve our understanding of pathogenetic mechanisms and patient stratification. Concurrently, the role of immune cells in the tumor microenvironment has received increasing attention, especially T cells and tumor-associated macrophages (TAM). The latter are a mixed population of activated brain-resident microglia and infiltrating monocytes/monocyte-derived macrophages, both of which express ionized calcium-binding adapter molecule 1 (IBA1). This study investigated differences in immune cell subpopulations among distinct transcriptional subtypes of GBM. Human GBM samples were molecularly characterized and assigned to Proneural, Mesenchymal or Classical subtypes as defined by NanoString nCounter Technology. Subsequently, we performed and analyzed automated immunohistochemical stainings for TAM as well as specific T cell populations. The Mesenchymal subtype of GBM showed the highest presence of TAM, CD8
+ , CD3+ and FOXP3+ T cells, as compared to Proneural and Classical subtypes. High expression levels of the TAM-related gene AIF1 , which encodes the TAM-specific protein IBA1, correlated with a worse prognosis in Proneural GBM, but conferred a survival benefit in Mesenchymal tumors. We used our data to construct a mathematical model that could reliably identify Mesenchymal GBM with high sensitivity using a combination of the aforementioned cell-specific IHC markers. In conclusion, we demonstrated that molecularly distinct GBM subtypes are characterized by profound differences in the composition of their immune microenvironment, which could potentially help to identify tumors amenable to immunotherapy., (© 2019 The Author(s). Published by Taylor & Francis.)- Published
- 2019
- Full Text
- View/download PDF
38. Arming oHSV with ULBP3 drives abscopal immunity in lymphocyte-depleted glioblastoma.
- Author
-
Wirsching HG, Zhang H, Szulzewsky F, Arora S, Grandi P, Cimino PJ, Amankulor N, Campbell JS, McFerrin L, Pattwell SS, Ene C, Hicks A, Ball M, Yan J, Zhang J, Kumasaka D, Pierce RH, Weller M, Finer M, Quéva C, Glorioso JC, Houghton AM, and Holland EC
- Subjects
- Animals, Antigen Presentation genetics, Antineoplastic Agents, Immunological pharmacology, Brain pathology, Brain Neoplasms genetics, Brain Neoplasms immunology, Brain Neoplasms mortality, Cell Line, Tumor, Combined Modality Therapy methods, Disease Models, Animal, Female, GPI-Linked Proteins genetics, GPI-Linked Proteins immunology, Gene Expression Regulation, Neoplastic immunology, Glioblastoma genetics, Glioblastoma immunology, Glioblastoma mortality, Humans, Intercellular Signaling Peptides and Proteins genetics, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase immunology, Kaplan-Meier Estimate, Male, Mice, Mice, Transgenic, Oncolytic Viruses genetics, Primary Cell Culture, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Recombinant Proteins genetics, Recombinant Proteins immunology, Simplexvirus genetics, Up-Regulation, Antineoplastic Agents, Immunological therapeutic use, Brain Neoplasms therapy, Glioblastoma therapy, Intercellular Signaling Peptides and Proteins immunology, Oncolytic Virotherapy methods, Oncolytic Viruses immunology, Simplexvirus immunology
- Abstract
Oncolytic viruses induce local tumor destruction and inflammation. Whether virotherapy can also overcome immunosuppression in noninfected tumor areas is under debate. To address this question, we have explored immunologic effects of oncolytic herpes simplex viruses (oHSVs) in a genetically engineered mouse model of isocitrate dehydrogenase (IDH) wild-type glioblastoma, the most common and most malignant primary brain tumor in adults. Our model recapitulates the genomics, the diffuse infiltrative growth pattern, and the extensive macrophage-dominant immunosuppression of human glioblastoma. Infection with an oHSV that was armed with a UL16-binding protein 3 (ULBP3) expression cassette inhibited distant tumor growth in the absence of viral spreading (abscopal effect) and yielded accumulation of activated macrophages and T cells. There was also abscopal synergism of oHSVULBP3 with anti-programmed cell death 1 (anti-PD-1) against distant, uninfected tumor areas; albeit consistent with clinical trials in patients with glioblastoma, monotherapy with anti-PD-1 was ineffective in our model. Arming oHSV with ULBP3 led to upregulation of antigen processing and presentation gene sets in myeloid cells. The cognate ULBP3 receptor NKG2D, however, is not present on myeloid cells, suggesting a noncanonical mechanism of action of ULBP3. Overall, the myeloid-dominant, anti-PD-1-sensitive abscopal effect of oHSVULBP3 warrants further investigation in patients with IDH wild-type glioblastoma.
- Published
- 2019
- Full Text
- View/download PDF
39. A De Novo Mouse Model of C11orf95-RELA Fusion-Driven Ependymoma Identifies Driver Functions in Addition to NF-κB.
- Author
-
Ozawa T, Arora S, Szulzewsky F, Juric-Sekhar G, Miyajima Y, Bolouri H, Yasui Y, Barber J, Kupp R, Dalton J, Jones TS, Nakada M, Kumabe T, Ellison DW, Gilbertson RJ, and Holland EC
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Transformation, Neoplastic, DNA-Binding Proteins metabolism, Disease Models, Animal, Ependymoma genetics, Ependymoma metabolism, Ependymoma pathology, Mice, Neural Stem Cells cytology, Neural Stem Cells metabolism, Oncogene Fusion, Principal Component Analysis, Signal Transduction, Transcription Factor RelA metabolism, Transcriptome, DNA-Binding Proteins genetics, Transcription Factor RelA genetics
- Abstract
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 (RELA
FUS ). 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., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
40. Increased HOXA5 expression provides a selective advantage for gain of whole chromosome 7 in IDH wild-type glioblastoma.
- Author
-
Cimino PJ, Kim Y, Wu HJ, Alexander J, Wirsching HG, Szulzewsky F, Pitter K, Ozawa T, Wang J, Vazquez J, Arora S, Rabadan R, Levine R, Michor F, and Holland EC
- Subjects
- Animals, Brain Neoplasms mortality, Brain Neoplasms pathology, Brain Neoplasms radiotherapy, Cell Proliferation, Chromosome Duplication, Glioblastoma mortality, Glioblastoma pathology, Glioblastoma radiotherapy, Homeodomain Proteins genetics, Humans, Isocitrate Dehydrogenase genetics, Mice, Neoplasm Recurrence, Local, Phosphoproteins genetics, Radiation Tolerance, Transcription Factors, Brain Neoplasms genetics, Chromosomes, Human, Pair 7, Glioblastoma genetics, Homeodomain Proteins metabolism, Phosphoproteins metabolism
- Abstract
Glioblastoma is the most frequently occurring and invariably fatal primary brain tumor in adults. The vast majority of glioblastomas is characterized by chromosomal copy number alterations, including gain of whole chromosome 7 and loss of whole chromosome 10. Gain of whole chromosome 7 is an early event in gliomagenesis that occurs in proneural-like precursor cells, which give rise to all isocitrate dehydrogenase (IDH) wild-type glioblastoma transcriptional subtypes. Platelet-derived growth factor A ( PDGFA ) is one gene on chromosome 7 known to drive gliomagenesis, but, given its location near the end of 7p, there are likely several other genes located along chromosome 7 that select for its increased whole-chromosome copy number within glioblastoma cells. To identify other potential genes that could select for gain of whole chromosome 7, we developed an unbiased bioinformatics approach that identified homeobox A5 ( HOXA5 ) as a gene whose expression correlated with gain of chromosome 7 and a more aggressive phenotype of the resulting glioma. High expression of HOXA5 in glioblastoma was associated with a proneural gene expression pattern and decreased overall survival in both human proneural and PDGF-driven mouse glioblastoma. Furthermore, HOXA5 overexpression promoted cellular proliferation and potentiated radioresistance. We also found enrichment of HOXA5 expression in recurrent human and mouse glioblastoma at first recurrence after radiotherapy. Overall, this study implicates HOXA5 as a chromosome 7-associated gene-level locus that promotes selection for gain of whole chromosome 7 and an aggressive phenotype in glioblastoma., (© 2018 Cimino et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
41. Loss of host-derived osteopontin creates a glioblastoma-promoting microenvironment.
- Author
-
Szulzewsky F, Schwendinger N, Güneykaya D, Cimino PJ, Hambardzumyan D, Synowitz M, Holland EC, and Kettenmann H
- Subjects
- Animals, Apoptosis, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Movement, Cell Proliferation, Disease Progression, Glioblastoma genetics, Glioblastoma metabolism, Humans, Mice, Mice, Inbred C57BL, Microglia metabolism, Monocytes metabolism, Osteopontin genetics, Brain Neoplasms pathology, Glioblastoma pathology, Microglia pathology, Monocytes pathology, Osteopontin metabolism, Tumor Microenvironment
- Abstract
Background: Microglia and periphery-derived monocytes infiltrate human and mouse glioblastoma and their density is positively correlated with malignancy. Using microarray and RNA sequencing, we have previously shown that glioblastoma-associated microglia/monocytes (GAMs) express osteopontin/SPP1., Methods: We used quantitative reverse transcriptase PCR, immunofluorescence stainings, western blot, and flow cytometry to identify the various sources of osteopontin (OPN) expression in human and mouse glioblastoma. We implanted wild type GL261 glioblastoma cells, which do not express significant levels of OPN, into wild type and OPN-/- mice to investigate the role of microenvironment-derived OPN on glioblastoma progression., Results: Our data indicate that GAMs are the predominant source of OPN in both human and mouse glioblastoma and express only the secreted form of OPN. Loss of microenvironment-derived OPN enhanced tumor progression. Staining by Ki67 and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling showed no difference in overall cell proliferation but a decreased apoptosis rate in tumors in OPN-/- mice. CD31 staining showed a significantly decreased number of microvessels in tumors in OPN-/- mice, accompanied by reduced coverage of vessels with platelet derived growth factor receptor β+ pericytes. Flow cytometry analysis revealed a significant increase of CD11b+/CD45low microglia but not of CD11b+/CD45high macrophages/monocytes in tumors in OPN-/- mice. Sorted CD11b+ cells from wild type and OPN-/- naïve brains and tumors did not show a significant difference in the expression pattern of activation marker genes., Conclusion: Our results show that in tested human and mouse glioblastoma samples, OPN is predominantly expressed and secreted by GAMs and that, in contrast to OPN expression in the tumor cells per se, loss of stroma-derived OPN creates a glioblastoma-promoting microenvironment., (© The Author(s) 2017. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com)
- Published
- 2018
- Full Text
- View/download PDF
42. Mutant IDH1 regulates the tumor-associated immune system in gliomas.
- Author
-
Amankulor NM, Kim Y, Arora S, Kargl J, Szulzewsky F, Hanke M, Margineantu DH, Rao A, Bolouri H, Delrow J, Hockenbery D, Houghton AM, and Holland EC
- Subjects
- Animals, Brain Neoplasms enzymology, Chemotaxis genetics, DNA Methylation, Disease Models, Animal, Glioma enzymology, Humans, Leukocyte Common Antigens metabolism, Leukocytes pathology, Mice, Mutation, Neutrophil Infiltration genetics, Neutrophils pathology, Brain Neoplasms genetics, Brain Neoplasms immunology, Glioma genetics, Glioma immunology, Immune System physiopathology, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase metabolism
- Abstract
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., (© 2017 Amankulor et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2017
- Full Text
- View/download PDF
43. Human glioblastoma-associated microglia/monocytes express a distinct RNA profile compared to human control and murine samples.
- Author
-
Szulzewsky F, Arora S, de Witte L, Ulas T, Markovic D, Schultze JL, Holland EC, Synowitz M, Wolf SA, and Kettenmann H
- Subjects
- Animals, CD11b Antigen metabolism, Computational Biology, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Mice, Real-Time Polymerase Chain Reaction, Sequence Analysis, RNA, Transcriptome, Brain Neoplasms metabolism, Glioblastoma metabolism, Microglia metabolism, Monocytes metabolism
- Abstract
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., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
44. The subpopulation of microglia expressing functional muscarinic acetylcholine receptors expands in stroke and Alzheimer's disease.
- Author
-
Pannell M, Meier MA, Szulzewsky F, Matyash V, Endres M, Kronenberg G, Prinz V, Waiczies S, Wolf SA, and Kettenmann H
- Subjects
- Animals, Brain metabolism, Carbachol pharmacology, Disease Models, Animal, Female, Glioma metabolism, Macrophages drug effects, Macrophages metabolism, Male, Mice, Models, Animal, Multiple Sclerosis metabolism, Muscarinic Agonists pharmacology, Neurotransmitter Agents metabolism, Receptor, Muscarinic M2 metabolism, Receptor, Muscarinic M3 metabolism, Alzheimer Disease metabolism, Microglia metabolism, Receptors, Muscarinic biosynthesis, Stroke metabolism
- Abstract
Microglia undergo a process of activation in pathology which is controlled by many factors including neurotransmitters. We found that a subpopulation (11 %) of freshly isolated adult microglia respond to the muscarinic acetylcholine receptor agonist carbachol with a Ca(2+) increase and a subpopulation of similar size (16 %) was observed by FACS analysis using an antibody against the M3 receptor subtype. The carbachol-sensitive population increased in microglia/brain macrophages isolated from tissue of mouse models for stroke (60 %) and Alzheimer's disease (25 %), but not for glioma and multiple sclerosis. Microglia cultured from adult and neonatal brain contained a carbachol-sensitive subpopulation (8 and 9 %), which was increased by treatment with interferon-γ to around 60 %. This increase was sensitive to blockers of protein synthesis and correlated with an upregulation of the M3 receptor subtype and with an increased expression of MHC-I and MHC-II. Carbachol was a chemoattractant for microglia and decreased their phagocytic activity.
- Published
- 2016
- Full Text
- View/download PDF
45. Loss of CX3CR1 increases accumulation of inflammatory monocytes and promotes gliomagenesis.
- Author
-
Feng X, Szulzewsky F, Yerevanian A, Chen Z, Heinzmann D, Rasmussen RD, Alvarez-Garcia V, Kim Y, Wang B, Tamagno I, Zhou H, Li X, Kettenmann H, Ransohoff RM, and Hambardzumyan D
- Subjects
- Animals, Brain Neoplasms metabolism, Brain Neoplasms pathology, CX3C Chemokine Receptor 1, Cell Line, Female, Gene Expression Regulation, Neoplastic, Glioblastoma metabolism, Glioblastoma pathology, Humans, Immunoblotting, Interleukin-1beta genetics, Interleukin-1beta metabolism, Interleukin-1beta pharmacology, Macrophages metabolism, Macrophages pathology, Male, Mice, Knockout, Mice, Transgenic, Microglia pathology, Microscopy, Confocal, Monocytes pathology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Receptors, Chemokine metabolism, Receptors, Interleukin-1 Type I genetics, Receptors, Interleukin-1 Type I metabolism, Reverse Transcriptase Polymerase Chain Reaction, Survival Analysis, Tumor Cells, Cultured, Tumor Microenvironment genetics, p38 Mitogen-Activated Protein Kinases metabolism, Brain Neoplasms genetics, Glioblastoma genetics, Microglia metabolism, Monocytes metabolism, Receptors, Chemokine genetics
- Abstract
The most abundant populations of non-neoplastic cells in the glioblastoma (GBM) microenvironment are resident microglia, macrophages and infiltrating monocytes from the blood circulation. The mechanisms by which monocytes infiltrate into GBM, their fate following infiltration, and their role in GBM growth are not known. Here we tested the hypothesis that loss of the fractalkine receptor CX3CR1 in microglia and monocytes would affect gliomagenesis. Deletion of Cx3cr1 from the microenvironment resulted in increased tumor incidence and shorter survival times in glioma-bearing mice. Loss of Cx3cr1 did not affect accumulation of microglia/macrophages in peri-tumoral areas, but instead indirectly promoted the trafficking of CD11b+CD45hiCX3CR1lowLy-6ChiLy-6G-F4/80-/low circulating inflammatory monocytes into the CNS, resulting in their increased accumulation in the perivascular area. Cx3cr1-deficient microglia/macrophages and monocytes demonstrated upregulation of IL1β expression that was inversely proportional to Cx3cr1 gene dosage. The Proneural subgroup of the TCGA GBM patient dataset with high IL1β expression showed shorter survival compared to patients with low IL1β. IL1β promoted tumor growth and increased the cancer stem cell phenotype in murine and human Proneural glioma stem cells (GSCs). IL1β activated the p38 MAPK signaling pathway and expression of monocyte chemoattractant protein (MCP-1/CCL2) by tumor cells. Loss of Cx3cr1 in microglia in a monocyte-free environment had no impact on tumor growth and did not alter microglial migration. These data suggest that enhancing signaling to CX3CR1 or inhibiting IL1β signaling in intra-tumoral macrophages can be considered as potential strategies to decrease the tumor-promoting effects of monocytes in Proneural GBM.
- Published
- 2015
- Full Text
- View/download PDF
46. Vascular signal transducer and activator of transcription-3 promotes angiogenesis and neuroplasticity long-term after stroke.
- Author
-
Hoffmann CJ, Harms U, Rex A, Szulzewsky F, Wolf SA, Grittner U, Lättig-Tünnemann G, Sendtner M, Kettenmann H, Dirnagl U, Endres M, and Harms C
- Subjects
- ADAM Proteins biosynthesis, ADAM Proteins genetics, ADAMTS9 Protein, Animals, Axons physiology, Brain pathology, Cellular Microenvironment, Cerebrovascular Circulation, Convalescence, Extracellular Matrix Proteins metabolism, Gene Expression Profiling, Infarction, Middle Cerebral Artery pathology, Mice, Mice, Knockout, Microglia pathology, Oligonucleotide Array Sequence Analysis, Phosphorylation, Protein Processing, Post-Translational, Recovery of Function, STAT3 Transcription Factor deficiency, STAT3 Transcription Factor genetics, Signal Transduction physiology, Endothelium, Vascular metabolism, Infarction, Middle Cerebral Artery physiopathology, Neovascularization, Physiologic physiology, Neuronal Plasticity physiology, STAT3 Transcription Factor physiology
- Abstract
Background: 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 Stat3(floxed/KO) and Tie2-Cre(ERT2) 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., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
47. Glioma-associated microglia/macrophages display an expression profile different from M1 and M2 polarization and highly express Gpnmb and Spp1.
- Author
-
Szulzewsky F, Pelz A, Feng X, Synowitz M, Markovic D, Langmann T, Holtman IR, Wang X, Eggen BJ, Boddeke HW, Hambardzumyan D, Wolf SA, and Kettenmann H
- Subjects
- Animals, Brain Neoplasms metabolism, Cell Line, Tumor, Cells, Cultured, Gene Expression Regulation, Neoplastic, Glioblastoma metabolism, Humans, Macrophages cytology, Membrane Glycoproteins metabolism, Mice, Microglia cytology, Neoplasm Transplantation, Osteopontin metabolism, Prognosis, Brain Neoplasms pathology, Glioblastoma pathology, Macrophages physiology, Membrane Glycoproteins genetics, Microglia physiology, Osteopontin genetics
- Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
48. Altered microglial phagocytosis in GPR34-deficient mice.
- Author
-
Preissler J, Grosche A, Lede V, Le Duc D, Krügel K, Matyash V, Szulzewsky F, Kallendrusch S, Immig K, Kettenmann H, Bechmann I, Schöneberg T, and Schulz A
- Subjects
- Animals, Brain cytology, CD11b Antigen genetics, CD11b Antigen metabolism, CX3C Chemokine Receptor 1, Cell Movement genetics, Flow Cytometry, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, In Vitro Techniques, Mice, Mice, Inbred C57BL, Mice, Transgenic, Organ Culture Techniques, Receptors, Chemokine genetics, Receptors, Chemokine metabolism, Receptors, Lysophospholipid genetics, Retina metabolism, Tumor Necrosis Factor-alpha metabolism, Gene Expression Regulation genetics, Microglia physiology, Phagocytosis genetics, Receptors, Lysophospholipid deficiency
- Abstract
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., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
49. The subpopulation of microglia sensitive to neurotransmitters/neurohormones is modulated by stimulation with LPS, interferon-γ, and IL-4.
- Author
-
Pannell M, Szulzewsky F, Matyash V, Wolf SA, and Kettenmann H
- Subjects
- Animals, Animals, Newborn, Cells, Cultured, Mice, Mice, Inbred C57BL, Interferon-gamma pharmacology, Interleukin-4 pharmacology, Lipopolysaccharides pharmacology, Microglia drug effects, Microglia metabolism, Neurotransmitter Agents pharmacology
- Abstract
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., (Copyright © 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
50. NTPDase1 activity attenuates microglial phagocytosis.
- Author
-
Bulavina L, Szulzewsky F, Rocha A, Krabbe G, Robson SC, Matyash V, and Kettenmann H
- Subjects
- Animals, Cells, Cultured, Immunohistochemistry, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Confocal, Organ Culture Techniques, Adenosine Triphosphate metabolism, Antigens, CD metabolism, Apyrase metabolism, Microglia metabolism, Phagocytosis physiology
- Abstract
Purinergic signaling plays a major role in the regulation of phagocytosis in microglia. Interplay between P2 and P1 receptor activation is controlled by a cascade of extracellular enzymes which dephosphorylate purines resulting in the formation of adenosine. The ATP- and ADP-degrading capacity of cultured microglia depends on the expression of ecto-nucleoside triphosphate diphosphohydrolase 1 (CD39) and is several times higher when compared to astrocytes which lack this enzyme. In brain slices, deletion of CD39 resulted in a 50 % decrease of ADP-degrading ability, while the degradation of ATP was decreased to about 75 % of the values measured in wild-type brain tissue. Microglia in acute slices from cd39(-/-) animals had increased constitutive phagocytic activity which could not be further enhanced by ATP in contrast to control animals. Pharmacological blockage of P2 receptors decreased the constitutive phagocytic activity to a similar base level in wild-type and cd39(-/-) microglia. Activation of P1 receptors by non-hydrolysable adenosine analog significantly decreased phagocytic activity. Deletion of CD73, an enzyme expressed by microglia which converts AMP to adenosine did not affect phagocytic activity. Taken together, these data show that CD39 plays a prominent role in controlling ATP levels and thereby microglial phagocytosis.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.