15 results on '"Anna-Eva Blank"'
Search Results
2. Cardiomyocyte Hyperplasia and Immaturity but not Hypertrophy are Characteristic Features of Patients with Rasopathies
- Author
-
Jörg-Detlef Drenckhahn, Luka Nicin, Sara Akhouaji, Svenja Krück, Anna Eva Blank, Anne Schänzer, Uygar Yörüker, Christian Jux, Wesley Abplanalp, David John, Andreas Zeiher, Stefanie Dimmeler, and Stefan Rupp
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
- Full Text
- View/download PDF
3. TGF-β activates pericytes via induction of the epithelial-to-mesenchymal transition protein SLUG in glioblastoma
- Author
-
Elena I. Ilina, Peter Baumgarten, Friedrich Feuerhake, Lisa Mäder, Kavi Devraj, Naita M. Wirsik, Michel Mittelbronn, Ulrike Naumann, Patrick N. Harter, Jakob Ehlers, Anna-Eva Blank, and Anne Grote
- Subjects
0301 basic medicine ,Histology ,Slug ,medicine.medical_treatment ,Motility ,Pathology and Forensic Medicine ,Receptor, Platelet-Derived Growth Factor beta ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Movement ,Transforming Growth Factor beta ,Physiology (medical) ,Glioma ,medicine ,Humans ,Epithelial–mesenchymal transition ,biology ,Chemistry ,Brain Neoplasms ,Endothelial Cells ,biology.organism_classification ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cytokine ,Neurology ,embryonic structures ,biology.protein ,Cancer research ,Neurology (clinical) ,Snail Family Transcription Factors ,Glioblastoma ,Pericytes ,030217 neurology & neurosurgery ,Platelet-derived growth factor receptor ,Transforming growth factor - Abstract
Aims In primary central nervous system tumours, epithelial-to-mesenchymal transition (EMT) gene expression is associated with increased malignancy. However, it has also been shown that EMT factors in gliomas are almost exclusively expressed by glioma vessel-associated pericytes (GA-Peris). In this study, we aimed to identify the mechanism of EMT in GA-Peris and its impact on angiogenic processes. Methods In glioma patients, vascular density and the expression of the pericytic markers platelet derived growth factor receptor (PDGFR)-β and smooth muscle actin (αSMA) were examined in relation to the expression of the EMT transcription factor SLUG and were correlated with survival of patients with glioblastoma (GBM). Functional mechanisms of SLUG regulation and the effects on primary human brain vascular pericytes (HBVP) were studied in vitro by measuring proliferation, cell motility and growth characteristics. Results The number of PDGFR-β- and αSMA-positive pericytes did not change with increased malignancy nor showed an association with the survival of GBM patients. However, SLUG-expressing pericytes displayed considerable morphological changes in GBM-associated vessels, and TGF-β induced SLUG upregulation led to enhanced proliferation, motility and altered growth patterns in HBVP. Downregulation of SLUG or addition of a TGF-β antagonising antibody abolished these effects. Conclusions We provide evidence that in GA-Peris, elevated SLUG expression is mediated by TGF-β, a cytokine secreted by most glioma cells, indicating that the latter actively modulate neovascularisation not only by modulating endothelial cells, but also by influencing pericytes. This process might be responsible for the formation of an unstructured tumour vasculature as well as for the breakdown of the blood-brain barrier in GBM.
- Published
- 2021
4. Distribution and prognostic impact of microglia/macrophage subpopulations in gliomas
- Author
-
Tony Kaoma, Pia S. Zeiner, Simone P. Niclou, Ane Iriondo, Kea Franz, Anna Golebiewska, Anne Grote, Monika Müller-Eschner, Friedrich Feuerhake, Arnaud Muller, Elena I. Ilina, Simon Bernatz, Patrick N. Harter, Katharina Filipski, Anna-Eva Blank, C. Preusse, Michel Mittelbronn, Werner Stenzel, Peter Baumgarten, Marcel A. Verhoff, Jenny Zinke, Jörg Wischhusen, Martin L. Hansmann, and Joachim P. Steinbach
- Subjects
0301 basic medicine ,education.field_of_study ,Tumor microenvironment ,Microglia ,CD68 ,General Neuroscience ,Population ,Biology ,medicine.disease ,nervous system diseases ,Pathology and Forensic Medicine ,MSR1 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,medicine.anatomical_structure ,Glioma ,medicine ,Cancer research ,Neurology (clinical) ,education ,CD163 ,030217 neurology & neurosurgery - Abstract
While the central nervous system is considered an immunoprivileged site and brain tumors display immunosuppressive features, both innate and adaptive immune responses affect glioblastoma (GBM) growth and treatment resistance. However, the impact of the major immune cell population in gliomas, represented by glioma-associated microglia/macrophages (GAMs), on patients' clinical course is still unclear. Thus, we aimed at assessing the immunohistochemical expression of selected microglia and macrophage markers in 344 gliomas (including gliomas from WHO grade I-IV). Furthermore, we analyzed a cohort of 241 IDH1R132H-non-mutant GBM patients for association of GAM subtypes and patient overall survival. Phenotypical properties of GAMs, isolated from high-grade astrocytomas by CD11b-based magnetic cell sorting, were analyzed by immunocytochemistry, mRNA microarray, qRT-PCR and bioinformatic analyses. A higher amount of CD68-, CD163- and CD206-positive GAMs in the vital tumor core was associated with beneficial patient survival. The mRNA expression profile of GAMs displayed an upregulation of factors that are considered as pro-inflammatory M1 (eg, CCL2, CCL3L3, CCL4, PTGS2) and anti-inflammatory M2 polarization markers (eg, MRC1, LGMN, CD163, IL10, MSR1), the latter rather being associated with phagocytic functions in the GBM microenvironment. In summary, we present evidence that human GBMs contain mixed M1/M2-like polarized GAMs and that the levels of different GAM subpopulations in the tumor core are positively associated with overall survival of patients with IDH1R132H-non-mutant GBMs.
- Published
- 2019
- Full Text
- View/download PDF
5. Pericytes/vessel-associated mural cells (VAMCs) are the major source of key epithelial-mesenchymal transition (EMT) factors SLUG and TWIST in human glioma
- Author
-
Michael Seifert, Barbara Klink, Peter Baumgarten, Simone P. Niclou, David Capper, Naita M. Wirsik, Stefan Liebner, Ulrike Naumann, Anna-Eva Blank, Michel Mittelbronn, Cornelia Zachskorn, Janina Jansong, Patrick N. Harter, Jakob Ehlers, and Lisa Mäder
- Subjects
0301 basic medicine ,Slug ,medicine.disease_cause ,pericytes ,Mural cell ,Malignant transformation ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Glioma ,medicine ,ddc:610 ,Epithelial–mesenchymal transition ,neoplasms ,Mutation ,biology ,EMT ,medicine.disease ,biology.organism_classification ,gliomas ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,embryonic structures ,MET ,Cancer research ,Immunohistochemistry ,Research Paper ,vessel-associated mural cells - Abstract
Epithelial-to-mesenchymal transition (EMT) is supposed to be responsible for increased invasion and metastases in epithelial cancer cells. The activation of EMT genes has further been proposed to be important in the process of malignant transformation of primary CNS tumors. Since the cellular source and clinical impact of EMT factors in primary CNS tumors still remain unclear, we aimed at deciphering their distribution in vivo and clinico-pathological relevance in human gliomas. We investigated 350 glioma patients for the expression of the key EMT factors SLUG and TWIST by immunohistochemistry and immunofluorescence related to morpho-genetic alterations such as EGFR-amplification, IDH-1 (R132H) mutation and 1p/19q LOH. Furthermore, transcriptional cluster and survival analyses were performed. Our data illustrate that SLUG and TWIST are overexpressed in gliomas showing vascular proliferation such as pilocytic astrocytomas and glioblastomas. EMT factors are exclusively expressed by non-neoplastic pericytes/vessel-associated mural cells (VAMCs). They are not associated with patient survival but correlate with pericytic/VAMC genes in glioblastoma cluster analysis. In summary, the upregulation of EMT genes in pilocytic astrocytomas and glioblastomas reflects the level of activation of pericytes/VAMCs in newly formed blood vessels. Our results underscore that the negative prognostic potential of the EMT signature in the group of diffuse gliomas of WHO grade II-IV does most likely not derive from glioma cells but rather reflects the degree of proliferating mural cells thereby constituting a potential target for future alternative treatment approaches.
- Published
- 2018
- Full Text
- View/download PDF
6. Differential expression of vascular endothelial growth factor A, its receptors VEGFR-1, -2, and -3 and co-receptors neuropilin-1 and -2 does not predict bevacizumab response in human astrocytomas
- Author
-
Peter Baumgarten, Benjamin Goeppert, Anna-Eva Blank, Joachim P. Steinbach, Katharina Hoffmann, Lisa Mäder, Elke Hattingen, Michel Mittelbronn, Patrick N. Harter, Oliver Bähr, Volker Seifert, Pia S. Zeiner, Kea Franz, Karl H. Plate, Marcia Machein, and Maika Dunst
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Angiogenesis Inhibitors ,Immunoenzyme Techniques ,0302 clinical medicine ,Neuropilin 1 ,Child ,Receptor ,Aged, 80 and over ,Brain Neoplasms ,Astrocytoma ,Middle Aged ,Prognosis ,Bevacizumab ,Survival Rate ,Vascular endothelial growth factor A ,Editorial ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,Adult ,Adolescent ,Young Adult ,03 medical and health sciences ,Glioma ,Biomarkers, Tumor ,medicine ,Humans ,Survival rate ,Aged ,Neoplasm Staging ,Vascular Endothelial Growth Factor Receptor-1 ,business.industry ,Growth factor ,Infant, Newborn ,Infant ,Vascular Endothelial Growth Factor Receptor-3 ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,Neuropilin-1 ,Neuropilin-2 ,Receptors, Vascular Endothelial Growth Factor ,030104 developmental biology ,Cancer research ,Neurology (clinical) ,Neoplasm Grading ,business ,Follow-Up Studies - Abstract
BACKGROUND A major hallmark of malignant progression in human astrocytomas is the formation of new blood vessels. Antiangiogenic therapy using the anti-vascular endothelial growth factor (VEGF)-antibody bevacizumab leads to increased progression-free survival in glioblastoma patients but does not influence their overall survival. To date, it is unclear why antiangiogenic therapy fails in many glioblastoma patients, while a small subpopulation profits considerably from this treatment. METHODS The aim of our study was to determine the expression of VEGF-A and its (co-) receptors by immunohistochemistry and to test the association with patient survival in 350 glioma patients. Additionally, VEGF-A expression was analyzed by in-situ hybridization. In 18 patients, the protein expression was compared with the bevacizumab response according to extended and modified RANO criteria. RESULTS We found a heterogeneous expression pattern of VEGF and its receptors in glioblastoma patients with significantly lower levels in WHO grade II and III tumors and normal-appearing brain tissue (P < .001). Pilocytic astrocytomas (WHO grade I) showed significantly higher VEGFR-1, -2 and neuropilin-1 levels as compared to WHO grade II and III astrocytomas (P < .01) but at lower levels than glioblastomas. The expression of neuropilin-2 was low in all tumors. There was neither a significant correlation between protein expression and patient survival nor between protein levels and bevacizumab response after modified RANO criteria. CONCLUSION Since our data indicate that beneficial response to bevacizumab treatment is independent of the expression of VEGF-A and its (co-) receptors, further investigation is needed to decipher the underlying mechanisms of antiangiogenic treatment response.
- Published
- 2015
- Full Text
- View/download PDF
7. Tumour necrosis factor receptor superfamily member 9 (TNFRSF9) is up-regulated in reactive astrocytes in human gliomas
- Author
-
Peter Baumgarten, Cornelia Zachskorn, Cathrin J. Czupalla, David Capper, Anna-Eva Blank, Christian Löffler, Patrick N. Harter, Michel Mittelbronn, Pia S. Zeiner, Karl H. Plate, and Jens Schittenhelm
- Subjects
Cell type ,Pathology ,medicine.medical_specialty ,Histology ,Microglia ,CD137 ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,medicine.anatomical_structure ,Neurology ,Downregulation and upregulation ,Gliosis ,Physiology (medical) ,Glioma ,medicine ,Immunohistochemistry ,Neurology (clinical) ,medicine.symptom ,Receptor - Abstract
Aims The prognosis of patients with malignant gliomas is still dismal despite maximum treatment. Novel therapeutic alternatives targeting tumorigenic pathways are, therefore, demanded. In murine glioma models, targeting of tumour necrosis factor receptor superfamily (TNFRSF) 9 led to complete tumour eradication. Thus, TNFRSF9 might also constitute a promising target in human diffuse gliomas. As there is a lack of data, we aimed to define the expression pattern and cellular source of TNFRSF9 in human gliomas. Methods We investigated TNFRSF9 expression in normal human central nervous system (CNS) tissue and glioma specimens using immunohistochemistry, immunofluorescence and Western blotting techniques. Results Our results show that TNFRSF9 is considerably up-regulated in human gliomas when compared with normal brain tissue. In addition, our data provides evidence for an immune cell-independent de novo expression pattern of TNFRSF9 in mainly non-neoplastic reactive astrocytes and excludes classic immunological cell types, namely lymphocytes and microglia as the source of TNFRSF9. Moreover, TNFRSF9 is predominantly expressed in a perivascular and peritumoural distribution with significantly higher expression in IDH-1 mutant gliomas. Conclusions Our findings provide a novel, TNFRSF9-positive, reactive astrocytic phenotype and challenge the therapeutic suitability of TNFRSF9 as a promising target for human gliomas.
- Published
- 2015
- Full Text
- View/download PDF
8. MIF Receptor CD74 is Restricted to Microglia/Macrophages, Associated with a M1-Polarized Immune Milieu and Prolonged Patient Survival in Gliomas
- Author
-
Jörg Wischhusen, Karl H. Plate, C. Preusse, Cornelia Zachskorn, Peter Baumgarten, Hansjürgen Bratzke, Ria Winkelmann, Jakob Weissenberger, Werner Stenzel, Christian Senft, Anna-Eva Blank, Anne K. Braczynski, Sandra Pennartz, Patrick N. Harter, Sandy Reiß, Michel Mittelbronn, Pia S. Zeiner, and Lixi Caspary
- Subjects
CD74 ,Microglia ,General Neuroscience ,Biology ,medicine.disease ,Molecular biology ,Pathology and Forensic Medicine ,Immune system ,medicine.anatomical_structure ,Glioma ,medicine ,biology.protein ,Immunohistochemistry ,Macrophage migration inhibitory factor ,Neurology (clinical) ,Antibody ,Receptor - Abstract
The macrophage migration inhibitory factor (MIF) receptor CD74 is overexpressed in various neoplasms, mainly in hematologic tumors, and currently investigated in clinical studies. CD74 is quickly internalized and recycles after antibody binding, therefore it constitutes an attractive target for antibody-based treatment strategies. CD74 has been further described as one of the most up-regulated molecules in human glioblastomas. To assess the potential relevance for anti-CD74 treatment, we determined the cellular source and clinicopathologic relevance of CD74 expression in human gliomas by immunohistochemistry, immunofluorescence, immunoblotting, cell sorting analysis and quantitative polymerase chain reaction (qPCR). Furthermore, we fractionated glioblastoma cells and glioma-associated microglia/macrophages (GAMs) from primary tumors and compared CD74 expression in cellular fractions with whole tumor lysates. Our results show that CD74 is restricted to GAMs in vivo, while being absent in tumor cells, the latter strongly expressing its ligand MIF. Most interestingly, a higher amount of CD74-positive GAMs was associated with beneficial patient survival constituting an independent prognostic parameter and with an anti-tumoral M1 polarization. In summary, CD74 expression in human gliomas is restricted to GAMs and positively associated with patient survival. In conclusion, CD74 represents a positive prognostic marker most probably because of its association with an M1-polarized immune milieu in high-grade gliomas.
- Published
- 2014
- Full Text
- View/download PDF
9. Loss of FUBP1 expression in gliomas predictsFUBP1mutation and is associated with oligodendroglial differentiation,IDH1mutation and 1p/19q loss of heterozygosity
- Author
-
David Capper, Tatjana Starzetz, Karl-Heinz Plate, Felix Sahm, B. Schwamb, Michel Mittelbronn, Venkatesh Kolluru, Patrick N. Harter, Martin Zörnig, Bernhard Radlwimmer, Martje Tönjes, U. Rabenhorst, Anna-Eva Blank, Peter Baumgarten, Ralf J. Rieker, Hiroko Ohgaki, A. von Deimling, and Donat Kögel
- Subjects
Mutation ,Histology ,IDH1 ,biology ,Cellular differentiation ,Cell cycle ,medicine.disease ,medicine.disease_cause ,Pathology and Forensic Medicine ,Loss of heterozygosity ,Neurology ,Physiology (medical) ,Ki-67 ,Glioma ,biology.protein ,medicine ,Cancer research ,Neurology (clinical) ,Oligodendroglioma - Abstract
Aims The Far Upstream Element [FUSE] Binding Protein 1 (FUBP1) regulates target genes, such as the cell cycle regulators MYC and p21. FUBP1 is up-regulated in many tumours and acts as an oncoprotein by stimulating proliferation and inhibiting apoptosis. Recently, FUBP1 mutations were identified in approximately 15% of oligodendrogliomas. To date, all reported FUBP1 mutations have been predicted to inactivate FUBP1, which suggests that in contrast to most other tumours FUBP1 may act as a tumour suppressor in oligodendrogliomas. Methods As no data are currently available concerning FUBP1 protein levels in gliomas, we examined the FUBP1 expression profiles of human glial tumours by immunohistochemistry and immunofluorescence. We analysed FUBP1 expression related to morphological differentiation, IDH1 and FUBP1 mutation status, 1p/19q loss of heterozygosity (LOH) as well as proliferation rate. Results Our findings demonstrate that FUBP1 expression levels are increased in all glioma subtypes as compared with normal central nervous system (CNS) control tissue and are associated with increased proliferation. In contrast, FUBP1 immunonegativity predicted FUBP1 mutation with a sensitivity of 100% and a specificity of 90% in our cohort and was associated with oligodendroglial differentiation, IDH1 mutation and 1p/19q loss of heterozygosity (LOH). Using this approach, we detected a to-date undescribed FUBP1 mutation in an oligodendroglioma. Conclusion In summary, our data indicate an association between of FUBP1 expression and proliferation in gliomas. Furthermore, our findings present FUBP1 immunohistochemical analysis as a helpful additional tool for neuropathological glioma diagnostics predicting FUBP1 mutation.
- Published
- 2014
- Full Text
- View/download PDF
10. ATP synthase deficiency due to TMEM70 mutation leads to ultrastructural mitochondrial degeneration and is amenable to treatment
- Author
-
Andreas Schulze, Werner Stenzel, Michel Mittelbronn, Patrick N. Harter, Matthias Kieslich, Anne K. Braczynski, Ulrich Drott, Klaus Müller, Rita Horvath, Karl H. Plate, Dominique S. Tews, Angela Abicht, Stefan Vlaho, Stephanie Kleinle, Anna-Eva Blank, Hans H. Goebel, Ilka Wittig, Horvath, Rita [0000-0002-9841-170X], Apollo - University of Cambridge Repository, and Mancuso, Michelangelo
- Subjects
500 Naturwissenschaften und Mathematik::570 Biowissenschaften ,Biologie ,Male ,medicine.medical_specialty ,Pathology ,Mitochondrial Diseases ,Article Subject ,Adolescent ,Cardiomyopathy ,lcsh:Medicine ,Oxidative phosphorylation ,Mitochondrion ,Biology ,medicine.disease_cause ,Mitochondrial Proton-Translocating ATPases ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Diagnosis, Differential ,Mitochondrial Proteins ,Internal medicine ,medicine ,Humans ,ddc:610 ,Child ,Gene ,Mutation ,General Immunology and Microbiology ,ATP synthase ,lcsh:R ,Membrane Proteins ,General Medicine ,medicine.disease ,Mitochondria ,Endocrinology ,Treatment Outcome ,Failure to thrive ,biology.protein ,Female ,medicine.symptom ,Research Article - Abstract
TMEM70 is involved in the biogenesis of mitochondrial ATP synthase and mutations in theTMEM70gene impair oxidative phosphorylation. Herein, we report on pathology and treatment of ATP synthase deficiency in four siblings. A consanguineous family of Roma (Gipsy) ethnic origin gave birth to 6 children of which 4 were affected presenting with dysmorphic features, failure to thrive, cardiomyopathy, metabolic crises, and 3-methylglutaconic aciduria as clinical symptoms. Genetic testing revealed a homozygous mutation (c.317-2A>G) in theTMEM70gene. While light microscopy was unremarkable, ultrastructural investigation of muscle tissue revealed accumulation of swollen degenerated mitochondria with lipid crystalloid inclusions, cristae aggregation, and exocytosis of mitochondrial material. Biochemical analysis of mitochondrial complexes showed an almost complete ATP synthase deficiency. Despite harbouring the same mutation, the clinical outcome in the four siblings was different. Two children died within 60 h after birth; the other two had recurrent life-threatening metabolic crises but were successfully managed with supplementation of anaplerotic amino acids, lipids, and symptomatic treatment during metabolic crisis. In summary,TMEM70mutations can cause distinct ultrastructural mitochondrial degeneration and almost complete deficiency of ATP synthase but are still amenable to treatment.
- Published
- 2015
11. MIF Receptor CD74 is Restricted to Microglia/Macrophages, Associated with a M1-Polarized Immune Milieu and Prolonged Patient Survival in Gliomas
- Author
-
Pia S, Zeiner, Corinna, Preusse, Anna-Eva, Blank, Cornelia, Zachskorn, Peter, Baumgarten, Lixi, Caspary, Anne K, Braczynski, Jakob, Weissenberger, Hansjürgen, Bratzke, Sandy, Reiß, Sandra, Pennartz, Ria, Winkelmann, Christian, Senft, Karl H, Plate, Jörg, Wischhusen, Werner, Stenzel, Patrick N, Harter, and Michel, Mittelbronn
- Subjects
Male ,Brain Neoplasms ,Macrophages ,Calcium-Binding Proteins ,Microfilament Proteins ,CD47 Antigen ,Glioma ,Kaplan-Meier Estimate ,Flow Cytometry ,Microarray Analysis ,Up-Regulation ,DNA-Binding Proteins ,Cell Line, Tumor ,Glial Fibrillary Acidic Protein ,Humans ,Female ,Microglia ,RNA, Messenger ,Research Articles - Abstract
The macrophage migration inhibitory factor (MIF) receptor CD74 is overexpressed in various neoplasms, mainly in hematologic tumors, and currently investigated in clinical studies. CD74 is quickly internalized and recycles after antibody binding, therefore it constitutes an attractive target for antibody‐based treatment strategies. CD74 has been further described as one of the most up‐regulated molecules in human glioblastomas. To assess the potential relevance for anti‐CD74 treatment, we determined the cellular source and clinicopathologic relevance of CD74 expression in human gliomas by immunohistochemistry, immunofluorescence, immunoblotting, cell sorting analysis and quantitative polymerase chain reaction (qPCR). Furthermore, we fractionated glioblastoma cells and glioma‐associated microglia/macrophages (GAMs) from primary tumors and compared CD74 expression in cellular fractions with whole tumor lysates. Our results show that CD74 is restricted to GAMs in vivo, while being absent in tumor cells, the latter strongly expressing its ligand MIF. Most interestingly, a higher amount of CD74‐positive GAMs was associated with beneficial patient survival constituting an independent prognostic parameter and with an anti‐tumoral M1 polarization. In summary, CD74 expression in human gliomas is restricted to GAMs and positively associated with patient survival. In conclusion, CD74 represents a positive prognostic marker most probably because of its association with an M1‐polarized immune milieu in high‐grade gliomas.
- Published
- 2014
12. Tumour necrosis factor receptor superfamily member 9 (TNFRSF9) is up-regulated in reactive astrocytes in human gliomas
- Author
-
Anna-Eva, Blank, Peter, Baumgarten, Pia, Zeiner, Cornelia, Zachskorn, Christian, Löffler, Jens, Schittenhelm, Cathrin J, Czupalla, David, Capper, Karl H, Plate, Patrick N, Harter, and Michel, Mittelbronn
- Subjects
Adult ,Aged, 80 and over ,Male ,Adolescent ,Brain Neoplasms ,Blotting, Western ,Infant, Newborn ,Fluorescent Antibody Technique ,Infant ,Glioma ,Middle Aged ,Immunohistochemistry ,Up-Regulation ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,Young Adult ,Astrocytes ,Child, Preschool ,Humans ,Female ,Child ,Aged - Abstract
The prognosis of patients with malignant gliomas is still dismal despite maximum treatment. Novel therapeutic alternatives targeting tumorigenic pathways are, therefore, demanded. In murine glioma models, targeting of tumour necrosis factor receptor superfamily (TNFRSF) 9 led to complete tumour eradication. Thus, TNFRSF9 might also constitute a promising target in human diffuse gliomas. As there is a lack of data, we aimed to define the expression pattern and cellular source of TNFRSF9 in human gliomas.We investigated TNFRSF9 expression in normal human central nervous system (CNS) tissue and glioma specimens using immunohistochemistry, immunofluorescence and Western blotting techniques.Our results show that TNFRSF9 is considerably up-regulated in human gliomas when compared with normal brain tissue. In addition, our data provides evidence for an immune cell-independent de novo expression pattern of TNFRSF9 in mainly non-neoplastic reactive astrocytes and excludes classic immunological cell types, namely lymphocytes and microglia as the source of TNFRSF9. Moreover, TNFRSF9 is predominantly expressed in a perivascular and peritumoural distribution with significantly higher expression in IDH-1 mutant gliomas.Our findings provide a novel, TNFRSF9-positive, reactive astrocytic phenotype and challenge the therapeutic suitability of TNFRSF9 as a promising target for human gliomas.
- Published
- 2013
13. Glioma cell migration and invasion as potential target for novel treatment strategies
- Author
-
Ulrike Naumann, Michel Mittelbronn, Patrick N. Harter, Anna-Eva Blank, Jennifer Rubel, Elena I. Ilina, and Hugo Bastida Esteban
- Subjects
Pathology ,medicine.medical_specialty ,General Neuroscience ,Biology ,Glioma cell ,medicine.disease ,Resection ,Extracellular matrix ,medicine.anatomical_structure ,Glioma ,medicine ,Treatment strategy ,Perivascular space ,Cytoskeleton ,Glioblastoma - Abstract
Diffuse human gliomas constitute a group of most treatment-refractory tumors even if maximum treatment strategies including neurosurgical resection followed by combined radio-/chemotherapy are applied. In contrast to most other neoplasms, diffusely infiltrating gliomas invade the brain along pre-existing structures such as axonal tracts and perivascular spaces. Even in cases of early diagnosis single or small clusters of glioma cells are already encountered far away from the main tumor bulk. Complex interactions between glioma cells and the surrounding extracellular matrix and considerable changes in the cytoskeletal apparatus are prerequisites for the cellular movement of glioma cells through the brain thereby escaping from most current treatments. This review provides an overview about classical and current concepts of glioma cell migration/invasion and promising preclinical treatment approaches.
- Published
- 2013
- Full Text
- View/download PDF
14. Fluid accumulation in the fetal extracerebral spaces: External hydrocephalus or subdural hygromas?
- Author
-
A Reitter, Patrick N. Harter, Anna-Eva Blank, Michel Mittelbronn, T Heinrich, and F Louwen
- Subjects
Fetus ,Pathology ,medicine.medical_specialty ,External hydrocephalus ,business.industry ,Pediatrics, Perinatology and Child Health ,Medicine ,Neurology (clinical) ,General Medicine ,Fluid accumulation ,business - Published
- 2011
- Full Text
- View/download PDF
15. Multifactorial modulation of susceptibility to l-lysine in an animal model of glutaric aciduria type I
- Author
-
Jürgen G. Okun, Shoko Komatsuzaki, David M. Koeller, Michel Mittelbronn, Anna Eva Blank, Sven W. Sauer, Stefan Kölker, Silvana Opp, and Peter Burgard
- Subjects
Male ,medicine.medical_specialty ,Mice, 129 Strain ,Metabolite ,Inborn errors of metabolism ,Biology ,Glutaric acid ,Neurodegenerative disease ,Aconitase ,Asymptomatic ,complex mixtures ,Glutarates ,chemistry.chemical_compound ,Sex Factors ,Risk Factors ,Internal medicine ,Glyceraldehyde ,medicine ,Hippocampus (mythology) ,Animals ,Genetic Predisposition to Disease ,Ketoglutarate Dehydrogenase Complex ,Animal model ,Molecular Biology ,Amino Acid Metabolism, Inborn Errors ,Aconitate Hydratase ,Mice, Knockout ,Dose-Response Relationship, Drug ,Glutaryl-CoA Dehydrogenase ,Brain Diseases, Metabolic ,Lysine ,Glutaric aciduria ,Brain ,Glyceraldehyde-3-Phosphate Dehydrogenases ,Neural metabolism ,medicine.disease ,Diet ,Amino acid ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,chemistry ,Spectrophotometry ,Molecular Medicine ,bacteria ,Female ,medicine.symptom ,Energy Metabolism ,Spongiosis - Abstract
Glutaric aciduria type I is an inherited defect in L-lysine, L-hydroxylysine and L-tryptophan degradation caused by deficiency of glutaryl-CoA dehydrogenase (GCDH). The majority of untreated patients presents with accumulation of neurotoxic metabolites - glutaric acid (GA) and 3-hydroxyglutaric acid (3-OHGA) - and striatal injury. Gcdh(-/-) mice display elevated levels of GA and 3-OH-GA but do not spontaneously develop striatal lesions. L-lysine-enriched diets (appr. 235 mg/d) were suggested to induce a neurological phenotype similar to affected patients. In our hands 93% of mice stressed according to the published protocol remained asymptomatic. To understand the underlying mechanism, we modified their genetic background (F1 C57BL6/Jx129/SvCrl) and increased the daily oral L-lysine supply (235-433 mg). We identified three modulating factors, (1) gender, (2) genetic background, and (3) amount of L-lysine. Male mice displayed higher vulnerability and inbreeding for more than two generations as well as elevating L-lysine supply increased the diet-induced mortality rate (up to 89%). Onset of first symptoms leads to strongly reduced intake of food and, thus, L-lysine suggesting a threshold for toxic metabolite production to induce neurological disease. GA and 3-OH-GA tissue concentrations did not correlate with dietary L-lysine supply but differed between symptomatic and asymptomatic mice. Cerebral activities of glyceraldehyde 3-phosphate dehydrogenase, 2-oxoglutarate dehydrogenase complex, and aconitase were decreased. Symptomatic mice did not develop striatal lesions or intracerebral hemorrhages. We found severe spongiosis in the hippocampus of Gcdh(-/-) mice which was independent of dietary L-lysine supply. In conclusion, the L-lysine-induced pathology in Gcdh(-/-) mice depends on genetic and dietary parameters.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.