16 results on '"Elena I. Ilina"'
Search Results
2. Linking epigenetic signature and metabolic phenotype in IDH mutant and IDH wildtype diffuse glioma
- Author
-
Elena I. Ilina, Jenny Zinke, Joachim P. Steinbach, Christian Senft, Patrick N. Harter, Bastian Roller, Karl H. Plate, Peter Baumgarten, Oliver Bähr, Katharina Filipski, Pia S. Zeiner, Simon Bernatz, Michael W. Ronellenfitsch, Elke Hattingen, Michel Mittelbronn, and Yannick Braun
- Subjects
0301 basic medicine ,Mitochondrial DNA ,Histology ,Lactate dehydrogenase A ,Biology ,Mitochondrion ,DNA, Mitochondrial ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Glioma ,medicine ,Humans ,Epigenetics ,education ,Gene ,education.field_of_study ,Brain Neoplasms ,DNA Methylation ,medicine.disease ,Isocitrate Dehydrogenase ,Phenotype ,030104 developmental biology ,Isocitrate dehydrogenase ,Neurology ,DNA methylation ,Cancer research ,Neurology (clinical) ,Transcriptome ,030217 neurology & neurosurgery - Abstract
Aims Changes in metabolism are known to contribute to tumour phenotypes. If and how metabolic alterations in brain tumours contribute to patient outcome is still poorly understood. Epigenetics impact metabolism and mitochondrial function. The aim of this study is a characterisation of metabolic features in molecular subgroups of isocitrate dehydrogenase mutant (IDHmut) and isocitrate dehydrogenase wildtype (IDHwt) gliomas. Methods We employed DNA methylation pattern analyses with a special focus on metabolic genes, large-scale metabolism panel immunohistochemistry (IHC), qPCR-based determination of mitochondrial DNA copy number and immune cell content using IHC and deconvolution of DNA methylation data. We analysed molecularly characterised gliomas (n = 57) for in depth DNA methylation, a cohort of primary and recurrent gliomas (n = 22) for mitochondrial copy number and validated these results in a large glioma cohort (n = 293). Finally, we investigated the potential of metabolic markers in Bevacizumab (Bev)-treated gliomas (n = 29). Results DNA methylation patterns of metabolic genes successfully distinguished the molecular subtypes of IDHmut and IDHwt gliomas. Promoter methylation of lactate dehydrogenase A negatively correlated with protein expression and was associated with IDHmut gliomas. Mitochondrial DNA copy number was increased in IDHmut tumours and did not change in recurrent tumours. Hierarchical clustering based on metabolism panel IHC revealed distinct subclasses of IDHmut and IDHwt gliomas with an impact on patient outcome. Further quantification of these markers allowed for the prediction of survival under anti-angiogenic therapy. Conclusion A mitochondrial signature was associated with increased survival in all analyses, which could indicate tumour subgroups with specific metabolic vulnerabilities.
- Published
- 2020
- Full Text
- View/download PDF
3. An Assessment of Mistletoe-Based Drugs Work in Synergy with Radio-Chemotherapy in the Treatment of Glioma in vitro and in vivo in Glioblastoma Bearing Mice
- Author
-
Sonja Schötterl, Andrea Gall, Elena I. Ilina, Naita M. Wirsik, Ulrike Naumann, Ingrid Ehrlich, Michel Mittelbronn, Stephan M. Huber, Jennifer T. Miemietz, and Hans Lentzen
- Subjects
Temozolomide ,biology ,business.industry ,medicine.medical_treatment ,medicine.disease ,biology.organism_classification ,In vivo ,Glioma ,Viscotoxins ,Adjuvant therapy ,Cancer research ,Viscum album ,Medicine ,business ,Cytotoxicity ,Adjuvant ,medicine.drug - Abstract
Extracts from the mistletoe plant Viscum album L. (VE) are used in the complementary cancer therapy in Europe for decades. VE contain several compounds like the mistletoe lectins (ML) 1-3 and viscotoxins (VT), but also several minor ingredients. ML-1 is suggested to be the main anti-tumor component of VE, VT induce cell death, and some minor compounds can strengthen the anti-tumor activity of ML-1. ML stimulate the immune system, induce cytotoxicity, are able to modify the expression of cancer-associated genes and influence the proliferation and motility of tumor cells. In this study we determine the anti-tumor effects of the VE ISCADOR Qu, of recombinant ML-1 (Aviscumine) and of native ML-1 in the treatment of glioblastoma (GBM), the most common brain tumor in adults. Additionally, we were determined whether a mistletoe-based therapy, if used as adjuvant treatment in combination with temozolomide(TMZ)-or irradiation, provide synergistic effects. The expression of the ML receptor CD75s, which is expressed in GBM specimen and cells, but not in the normal brain, correlates well with the drug-induced cytotoxicity. In GBM cells, the drugs induce cell death in a concentration-dependent manner, induce an arrest in the G2/M phase and provide synergistic and additive anti-tumor effects. A single intratumoral application of Aviscumine prolonged the survival of GBM mice longer than tumor irradiation and was even better if used in combination with radio-chemotherapy. This indicates that an adjuvant treatment of glioma patients with ML-containing drugs might be beneficial.
- Published
- 2021
- Full Text
- View/download PDF
4. 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
5. Enzymatic Activity of Glycosyltransferase Glt8d1 Promotes Human Glioblastoma Cells Migration
- Author
-
Tanja R. Müller, Monika Dieterle, Jean-Jacques Gérardy, Anne Schuster, Michel Mittelbronn, Elena I. Ilina, Céline Jeanty, Maitane Duarte Garcia-Escudero, Camille Cialini, Simone P. Niclou, Alina Moter, Gunnar Dittmar, Dietlind L. Gerloff, Vincent Puard, and Anna Golebiewska
- Subjects
biology ,Chemistry ,Brain tumor ,Cell migration ,medicine.disease ,In vitro ,Cell culture ,Glioma ,Glycosyltransferase ,Parenchyma ,biology.protein ,medicine ,Cancer research ,Cytoskeleton - Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor characterized by infiltrative migration of malignant glioma cells into the surrounding brain parenchyma. In this study, our analysis of GBM patient cohorts displayed significant higher expression of Glycosyltransferase 8 domain containing 1 ( GLT8D1 ) compared to normal brain tissue and could be associated with impaired patient survival. In vitro an increased GLT8D1 expression significantly enhanced migration of two different sphere-forming GBM cell lines. By in silico analysis we predicted the 3D-structure as well as the active site residues of GLT8D1. The introduction of point mutations in predicted active site reduced its glycosyltransferase activity in vitro and consequently impaired GBM tumor cell migration. Examination of GLT8D1 interaction partners by LC-MS/MS implied proteins associated with cytoskeleton and intracellular transport as potential substrates. In conclusion, we demonstrated that the enzymatic activity of glycosyltransferase GLT8D1 promotes GBM cell migration.
- Published
- 2021
- Full Text
- View/download PDF
6. 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
7. Enzymatic activity of glycosyltransferase GLT8D1 promotes human glioblastoma cell migration
- Author
-
Elena I. Ilina, Camille Cialini, Dietlind L. Gerloff, Maitane Duarte Garcia-Escudero, Céline Jeanty, Marie-Laëtitia Thézénas, Antoine Lesur, Vincent Puard, François Bernardin, Alina Moter, Anne Schuster, Monika Dieterle, Anna Golebiewska, Jean-Jacques Gérardy, Gunnar Dittmar, Simone P. Niclou, Tanja Müller, and Michel Mittelbronn
- Subjects
Cell biology ,Multidisciplinary ,Science ,Glycobiology ,Biochemistry ,Cancer - Abstract
Summary: Glioblastoma (GBM) is the most aggressive primary brain tumor characterized by infiltrative growth of malignant glioma cells into the surrounding brain parenchyma. In this study, our analysis of GBM patient cohorts revealed a significantly higher expression of Glycosyltransferase 8 domain containing 1 (GLT8D1) compared to normal brain tissue and could be associated with impaired patient survival. Increased in vitro expression of GLT8D1 significantly enhanced migration of two different sphere-forming GBM cell lines. By in silico analysis we predicted the 3D-structure as well as the active site residues of GLT8D1. The introduction of point mutations in the predicted active site reduced its glycosyltransferase activity in vitro and consequently impaired GBM tumor cell migration. Examination of GLT8D1 interaction partners by LC-MS/MS implied proteins associated with cytoskeleton and intracellular transport as potential substrates. In conclusion, we demonstrated that the enzymatic activity of glycosyltransferase GLT8D1 promotes GBM cell migration.
- Published
- 2022
- Full Text
- View/download PDF
8. Carboxypeptidase E transmits its anti-migratory function in glioma cells via transcriptional regulation of cell architecture and motility regulating factors
- Author
-
Tony Kaoma, Elena I. Ilina, Simone P. Niclou, Arnaud Muller, Michel Mittelbronn, Ulrike Naumann, Marcel A. Krüger, Laurent Vallar, and Angela Armento
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Motility ,CDC42 ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Animals ,Humans ,Neoplasm Invasiveness ,RNA, Messenger ,Regulation of gene expression ,Cell growth ,Gene Expression Profiling ,Carboxypeptidase H ,Cell migration ,Cell cycle ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Carboxypeptidase E ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Glioblastoma ,Signal Transduction - Abstract
Glioblastoma (GBM), the most frequent and aggressive malignant primary brain tumor, is characterized by a highly invasive growth. In our previous study we showed that overexpression of Carboxypeptidase E (CPE) mitigated glioma cell migration. In the present study we aimed at deciphering the regulatory mechanisms of the secreted form of CPE (sCPE). By transcriptome analysis and inhibition of signaling pathways involved in the regulation of cell growth and motility, we discovered that overexpression of sCPE was accompanied by differential regulation of mRNAs connected to the motility-associated networks, among others FAK, PAK, Cdc42, integrin, STAT3 as well as TGF-β. Especially SLUG was downregulated in sCPE-overexpressing glioma cells, paralleled by reduced expression of matrix-metalloproteinases (MMP) and, in consequence, by decreased cell migration. Expression of SLUG was regulated by ERK since inhibition of ERK reverted sCPE-mediated SLUG downregulation and enhanced cell motility. In a mouse glioma model, overexpression of sCPE significantly prolonged survival. Our results implicate a novel role for sCPE that mainly affects the expression of motility-associated genes via several signal pathways.
- Published
- 2017
- Full Text
- View/download PDF
9. Impact of Docetaxel on blood-brain barrier function and formation of breast cancer brain metastases
- Author
-
Bernhard C. Pestalozzi, Christoph Renner, Sascha Kleber, Frits Thorsen, Christine Solbach, Lukas Jennewein, Patrick N. Harter, Axel Mischo, Klaus Mueller, Kavi Devraj, Yannick Braun, Cornelia Penski, Rashi Halder, Elena I. Ilina, Simon Bernatz, Michael Mittelbronn, University of Zurich, and Mischo, Axel
- Subjects
0301 basic medicine ,Cancer Research ,Docetaxel ,Mice ,0302 clinical medicine ,Breast cancer ,Tubulin ,1306 Cancer Research ,Claudin-5 ,Brain Neoplasms ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Magnetic Resonance Imaging ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Blood-Brain Barrier ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,2730 Oncology ,medicine.drug ,Antineoplastic Agents ,Breast Neoplasms ,610 Medicine & health ,Taxane ,Blood–brain barrier ,lcsh:RC254-282 ,03 medical and health sciences ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Animals ,Humans ,ddc:610 ,TEER ,business.industry ,Sequence Analysis, RNA ,Gene Expression Profiling ,Research ,Brain metastasis ,medicine.disease ,Xenograft Model Antitumor Assays ,Microscopy, Electron ,030104 developmental biology ,Apoptosis ,10032 Clinic for Oncology and Hematology ,Cancer research ,business ,BBB - Abstract
Background Breast cancer (BC) is the most frequent malignant tumor in females and the 2nd most common cause of brain metastasis (BM), that are associated with a fatal prognosis. The increasing incidence from 10% up to 40% is due to more effective treatments of extracerebral sites with improved prognosis and increasing use of MRI in diagnostics. A frequently administered, potent chemotherapeutic group of drugs for BC treatment are taxanes usually used in the adjuvant and metastatic setting, which, however, have been suspected to be associated with a higher incidence of BM. The aim of our study was to experimentally analyze the impact of the taxane docetaxel (DTX) on brain metastasis formation, and to elucidate the underlying molecular mechanism. Methods A monocentric patient cohort was analyzed to determine the association of taxane treatment and BM formation. To identify the specific impact of DTX, a murine brain metastatic model upon intracardial injection of breast cancer cells was conducted. To approach the functional mechanism, dynamic contrast-enhanced MRI and electron microscopy of mice as well as in-vitro transendothelial electrical resistance (TEER) and tracer permeability assays using brain endothelial cells (EC) were carried out. PCR-based, immunohistochemical and immunoblotting analyses with additional RNA sequencing of murine and human ECs were performed to explore the molecular mechanisms by DTX treatment. Results Taxane treatment was associated with an increased rate of BM formation in the patient cohort and the murine metastatic model. Functional studies did not show unequivocal alterations of blood-brain barrier properties upon DTX treatment in-vivo, but in-vitro assays revealed a temporary DTX-related barrier disruption. We found disturbance of tubulin structure and upregulation of tight junction marker claudin-5 in ECs. Furthermore, upregulation of several members of the tubulin family and downregulation of tetraspanin-2 in both, murine and human ECs, was induced. Conclusion In summary, a higher incidence of BM was associated with prior taxane treatment in both a patient cohort and a murine mouse model. We could identify tubulin family members and tetraspanin-2 as potential contributors for the destabilization of the blood-brain barrier. Further analyses are needed to decipher the exact role of those alterations on tumor metastatic processes in the brain.
- Published
- 2019
- Full Text
- View/download PDF
10. Mistletoe-based drugs work in synergy with radio-chemotherapy in the treatment of glioma in vitro and in vivo in glioblastoma bearing mice
- Author
-
Hans Lentzen, Sonja Schötterl, Andrea Gall, Elena I. Ilina, Naita M. Wirsik, Jennifer T. Miemietz, Stephan M. Huber, Ulrike Naumann, Ingrid Ehrlich, Michel Mittelbronn, and Urech, Konrad
- Subjects
Article Subject ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Glioma ,Viscotoxins ,medicine ,Viscum album ,ddc:610 ,030304 developmental biology ,0303 health sciences ,biology ,Cell growth ,business.industry ,lcsh:Other systems of medicine ,Cell cycle ,lcsh:RZ201-999 ,medicine.disease ,biology.organism_classification ,Complementary and alternative medicine ,030220 oncology & carcinogenesis ,Cancer research ,business ,Cell culture assays ,Ex vivo ,Research Article - Abstract
Background. Extracts from Viscum album L. (VE) are used in the complementary cancer therapy in Europe for decades. VE contain several compounds like the mistletoe lectins (MLs) 1-3 and viscotoxins and also several minor ingredients. Since mistletoe lectin 1 (ML-1) has been described as the main component of VE harboring antitumor activity, purified native or recombinant ML-1 has been recently used in clinical trials. MLs stimulate the immune system, induce cytotoxicity, are able to modify the expression of cancer-associated genes, and influence the proliferation and motility of tumor cells. Objective. In this study our goal was to determine anticancer effects of the VE ISCADOR Qu, of recombinant ML-1 (Aviscumine), and of native ML-1 in the treatment of glioblastoma (GBM), the most common and highly malignant brain tumor in adults. Additionally we were interested whether these drugs, used in combination with a temozolomide-(TMZ)-based radio-chemotherapy, provide synergistic effects. Methods. Cell culture assays, ex vivo murine hippocampal brain slice cultures, human GBM cryosections, and a xenograft orthotopic glioblastoma mouse model were used. Results. In cells, the expression of the ML receptor CD75s, which is also expressed in GBM specimen, but not in normal brain, correlates with the drug-induced cytotoxicity. In GBM cells, the drugs induce cell death in a concentration-dependent manner and reduce cell growth by inducing cell cycle arrest in the G2/M phase. The cell cycle arrest was paralleled by modifications in the expression of cell cycle regulating genes. ML containing drugs, if combined with glioma standard therapy, provide synergistic and additive anticancer effects. Despite not reaching statistical significance, a single intratumoral application of Aviscumine prolonged the median survival of GBM mice longer than tumor irradiation. Moreover, intratumorally applied Aviscumine prolonged the survival of GBM-bearing mice if used in combination with irradiation and TMZ for further 6.5 days compared to the radio-chemotherapy. Conclusion. Our results suggest that an adjuvant treatment of glioma patients with ML-containing drugs might be beneficial.
- Published
- 2019
11. Effects of soluble CPE on glioma cell migration are associated with mTOR activation and enhanced glucose flux
- Author
-
Lukas Jennewein, Patrick N. Harter, Sven Zukunft, Michael W. Ronellenfitsch, Elena I. Ilina, Michel Mittelbronn, Dorothea Schulte, Ingrid Fleming, Ulrike Naumann, Marina Reichlmeir, David Capper, Leticia Sanchez, Cornelia Penski, Christian Behrends, Francois Le Guerroue, Yannick Braun, Felix Sahm, and Angela Armento
- Subjects
0301 basic medicine ,Motility ,RAC1 ,migration ,03 medical and health sciences ,Glioma ,Medicine ,ddc:610 ,PI3K/AKT/mTOR pathway ,biology ,business.industry ,glioblastoma ,Cell migration ,CPE ,medicine.disease ,Cell biology ,030104 developmental biology ,Oncology ,Carboxypeptidase E ,Anaerobic glycolysis ,mTOR ,biology.protein ,Phosphorylation ,business ,metabolism ,Research Paper - Abstract
Carboxypeptidase E (CPE) has recently been described as a multifunctional protein that regulates proliferation, migration and survival in several tumor entities. In glioblastoma (GBM), the most malignant primary brain tumor, secreted CPE (sCPE) was shown to modulate tumor cell migration. In our current study, we aimed at clarifying the underlying molecular mechanisms regulating anti-migratory as well as novel metabolic effects of sCPE in GBM. Here we show that sCPE activates mTORC1 signaling in glioma cells detectable by phosphorylation of its downstream target RPS6. Additionally, sCPE diminishes glioma cell migration associated with a negative regulation of Rac1 signaling via RPS6, since both inhibition of mTOR and stimulation of Rac1 results in a reversed effect of sCPE on migration. Knockdown of CPE leads to a decrease of active RPS6 associated with increased GBM cell motility. Apart from this, we show that sCPE enhances glucose flux into the tricarboxylic acid cycle at the expense of lactate production, thereby decreasing aerobic glycolysis, which might as well contribute to a less invasive behavior of tumor cells. Our data contributes to a better understanding of the complexity of GBM cell migration and sheds new light on how tumor cell invasion and metabolic plasticity are interconnected.
- Published
- 2017
12. Dominant-negative effects ofKCNQ2mutations are associated with epileptic encephalopathy
- Author
-
Elke Guenther, Elena I. Ilina, Timm Danker, Snezana Maljevic, Stephanie Nadine Reichel, Nicole Jezutkovic, Merle Bock, Simone Mandelstam, Heidi Löffler, Arvid Suls, Gökce Orhan, Ingrid E. Scheffer, Holger Lerche, Sarah Weckhuysen, Peter De Jonghe, and Dorien Schepers
- Subjects
Genetics ,medicine.medical_specialty ,Mutation ,Chemistry ,Voltage clamp ,Retigabine ,medicine.disease_cause ,medicine.disease ,Potassium channel ,Epilepsy ,chemistry.chemical_compound ,Endocrinology ,Neurology ,Internal medicine ,medicine ,Benign familial neonatal seizures ,Neurology (clinical) ,Cognitive decline ,Haploinsufficiency - Abstract
Objective Mutations in KCNQ2 and KCNQ3, encoding the voltage-gated potassium channels KV7.2 and KV7.3, are known to cause benign familial neonatal seizures mainly by haploinsufficiency. Here, we set out to determine the disease mechanism of 7 de novo missense KCNQ2 mutations that were recently described in patients with a severe epileptic encephalopathy including pharmacoresistant seizures and pronounced intellectual disability. Methods Mutations were inserted into the KCNQ2 cDNA. Potassium currents were recorded using 2-microelectrode voltage clamping, and surface expression was analyzed by a biotinylation assay in cRNA-injected Xenopus laevis oocytes. Results We observed a clear loss of function for all mutations. Strikingly, 5 of 7 mutations exhibited a drastic dominant-negative effect on wild-type KV7.2 or KV7.3 subunits, either by globally reducing current amplitudes (3 pore mutations) or by a depolarizing shift of the activation curve (2 voltage sensor mutations) decreasing potassium currents at the subthreshold level at which these channels are known to critically influence neuronal firing. One mutation significantly reduced surface expression. Application of retigabine, a recently marketed KV7 channel opener, partially reversed these effects for the majority of analyzed mutations. Interpretation The development of severe epilepsy and cognitive decline in children carrying 5 of the 7 studied KCNQ2 mutations can be related to a dominant-negative reduction of the resulting potassium current at subthreshold membrane potentials. Other factors such as genetic modifiers have to be postulated for the remaining 2 mutations. Retigabine or similar drugs may be used as a personalized therapy for this severe disease. Ann Neurol 2014;75:382–394
- Published
- 2014
- Full Text
- View/download PDF
13. Immunohistochemical Assessment of Phosphorylated mTORC1-Pathway Proteins in Human Brain Tumors
- Author
-
Elena I. Ilina, Peter Baumgarten, Michael C. Burger, Joachim P. Steinbach, Patrick N. Harter, Christian Senft, Julia Tichy, Martin Zörnig, Anna-Luisa Thiepold, Michel Mittelbronn, Michael W. Ronellenfitsch, and Lukas Jennewein
- Subjects
Pathology ,medicine.medical_specialty ,Immunocytochemistry ,Brain tumor ,lcsh:Medicine ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Glioma ,Cell Line, Tumor ,medicine ,Humans ,Phosphorylation ,lcsh:Science ,Multidisciplinary ,Chemistry ,Brain Neoplasms ,TOR Serine-Threonine Kinases ,lcsh:R ,medicine.disease ,Immunohistochemistry ,Staining ,Cell culture ,Multiprotein Complexes ,Cancer cell ,lcsh:Q ,Glioblastoma ,Research Article ,Signal Transduction - Abstract
Background: Current pathological diagnostics include the analysis of (epi-)genetic alterations as well as oncogenic pathways. Deregulated mammalian target of rapamycin complex 1 (mTORC1) signaling has been implicated in a variety of cancers including malignant gliomas and is considered a promising target in cancer treatment. Monitoring of mTORC1 activity before and during inhibitor therapy is essential. The aim of our study is to provide a recommendation and report on pitfalls in the use of phospho-specific antibodies against mTORC1-targets phospho-RPS6 (Ser235/236; Ser240/244) and phospho-4EBP1 (Thr37/46) in formalin fixed, paraffin embedded material. Methods and findings: Primary, established cell lines and brain tumor tissue from routine diagnostics were assessed by immunocyto-, immunohistochemistry, immunofluorescent stainings and immunoblotting. For validation of results, immunoblotting experiments were performed. mTORC-pathway activation was pharmacologically inhibited by torin2 and rapamycin. Torin2 treatment led to a strong reduction of signal intensity and frequency of all tested antibodies. In contrast phospho-4EBP1 did not show considerable reduction in staining intensity after rapamycin treatment, while immunocytochemistry with both phospho-RPS6-specific antibodies showed a reduced signal compared to controls. Staining intensity of both phospho-RPS6-specific antibodies did not show considerable decrease in stability in a timeline from 0–230 minutes without tissue fixation, however we observed a strong decrease of staining intensity in phospho-4EBP1 after 30 minutes. Detection of phospho-signals was strongly dependent on tissue size and fixation gradient. mTORC1-signaling was significantly induced in glioblastomas although not restricted to cancer cells but also detectable in non-neoplastic cells. Conclusion: Here we provide a recommendation for phospho-specific immunohistochemistry for patient-orientated therapy decisions and monitoring treatment response.
- Published
- 2015
14. Dominant-negative effects of KCNQ2 mutations are associated with epileptic encephalopathy
- Author
-
Gökce, Orhan, Merle, Bock, Dorien, Schepers, Elena I, Ilina, Stephanie Nadine, Reichel, Heidi, Löffler, Nicole, Jezutkovic, Sarah, Weckhuysen, Simone, Mandelstam, Arvid, Suls, Timm, Danker, Elke, Guenther, Ingrid E, Scheffer, Peter, De Jonghe, Holger, Lerche, and Snezana, Maljevic
- Subjects
Potassium Channels, Voltage-Gated ,Xenopus ,Mutation, Missense ,Oocytes ,Animals ,Humans ,KCNQ2 Potassium Channel ,Genetic Predisposition to Disease ,Carbamates ,Phenylenediamines ,Epilepsy, Benign Neonatal ,Membrane Potentials - Abstract
Mutations in KCNQ2 and KCNQ3, encoding the voltage-gated potassium channels KV 7.2 and KV 7.3, are known to cause benign familial neonatal seizures mainly by haploinsufficiency. Here, we set out to determine the disease mechanism of 7 de novo missense KCNQ2 mutations that were recently described in patients with a severe epileptic encephalopathy including pharmacoresistant seizures and pronounced intellectual disability.Mutations were inserted into the KCNQ2 cDNA. Potassium currents were recorded using 2-microelectrode voltage clamping, and surface expression was analyzed by a biotinylation assay in cRNA-injected Xenopus laevis oocytes.We observed a clear loss of function for all mutations. Strikingly, 5 of 7 mutations exhibited a drastic dominant-negative effect on wild-type KV 7.2 or KV 7.3 subunits, either by globally reducing current amplitudes (3 pore mutations) or by a depolarizing shift of the activation curve (2 voltage sensor mutations) decreasing potassium currents at the subthreshold level at which these channels are known to critically influence neuronal firing. One mutation significantly reduced surface expression. Application of retigabine, a recently marketed KV 7 channel opener, partially reversed these effects for the majority of analyzed mutations.The development of severe epilepsy and cognitive decline in children carrying 5 of the 7 studied KCNQ2 mutations can be related to a dominant-negative reduction of the resulting potassium current at subthreshold membrane potentials. Other factors such as genetic modifiers have to be postulated for the remaining 2 mutations. Retigabine or similar drugs may be used as a personalized therapy for this severe disease.
- Published
- 2013
15. 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
16. A hereditary spastic paraplegia mouse model supports a role of ZFYVE26/SPASTIZIN for the endolysosomal system
- Author
-
Ludger Schöls, Christian A. Hübner, Sandor Nietzsche, Christoph Biskup, Elena I. Ilina, J. Christopher Hennings, Antje K. Huebner, Amir Jahic, Judit Symmank, Christian Beetz, Thomas Braulke, Michael M. Kessels, Nicole Koch, Katrin Kollmann, Ingo Kurth, Kathrin N. Karle, Britta Qualmann, Mukhran Khundadze, and Geraldine Zimmer
- Subjects
Retinal degeneration ,Cancer Research ,Cerebellum ,pathology [Retinal Degeneration] ,pathology [Spastic Paraplegia, Hereditary] ,genetics [Retinal Degeneration] ,spastizin protein, human ,genetics [Carrier Proteins] ,genetics [Lysosomes] ,Corpus Callosum ,metabolism [Lysosomes] ,Mice ,metabolism [Corpus Callosum] ,pathology [Brain] ,Genetics (clinical) ,Motor Neurons ,Mice, Knockout ,LAMP1 ,metabolism [Retinal Degeneration] ,Retinal Degeneration ,Brain ,Anatomy ,pathology [Corpus Callosum] ,Cell biology ,medicine.anatomical_structure ,Knockout mouse ,medicine.symptom ,Research Article ,metabolism [Endosomes] ,lcsh:QH426-470 ,Neurite ,Hereditary spastic paraplegia ,Endosomes ,Biology ,genetics [Spastic Paraplegia, Hereditary] ,Genetics ,medicine ,Animals ,Humans ,ddc:610 ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,pathology [Endosomes] ,Cerebellar ataxia ,Spastic Paraplegia, Hereditary ,metabolism [Motor Neurons] ,medicine.disease ,lcsh:Genetics ,Disease Models, Animal ,nervous system ,Membrane protein ,metabolism [Brain] ,metabolism [Spastic Paraplegia, Hereditary] ,Mutation ,Lysosomes ,Carrier Proteins ,metabolism [Carrier Proteins] - Abstract
Hereditary spastic paraplegias (HSPs) are characterized by progressive weakness and spasticity of the legs because of the degeneration of cortical motoneuron axons. SPG15 is a recessively inherited HSP variant caused by mutations in the ZFYVE26 gene and is additionally characterized by cerebellar ataxia, mental decline, and progressive thinning of the corpus callosum. ZFYVE26 encodes the FYVE domain-containing protein ZFYVE26/SPASTIZIN, which has been suggested to be associated with the newly discovered adaptor protein 5 (AP5) complex. We show that Zfyve26 is broadly expressed in neurons, associates with intracellular vesicles immunopositive for the early endosomal marker EEA1, and co-fractionates with a component of the AP5 complex. As the function of ZFYVE26 in neurons was largely unknown, we disrupted Zfyve26 in mice. Zfyve26 knockout mice do not show developmental defects but develop late-onset spastic paraplegia with cerebellar ataxia confirming that SPG15 is caused by ZFYVE26 deficiency. The morphological analysis reveals axon degeneration and progressive loss of both cortical motoneurons and Purkinje cells in the cerebellum. Importantly, neuron loss is preceded by accumulation of large intraneuronal deposits of membrane-surrounded material, which co-stains with the lysosomal marker Lamp1. A density gradient analysis of brain lysates shows an increase of Lamp1-positive membrane compartments with higher densities in Zfyve26 knockout mice. Increased levels of lysosomal enzymes in brains of aged knockout mice further support an alteration of the lysosomal compartment upon disruption of Zfyve26. We propose that SPG15 is caused by an endolysosomal membrane trafficking defect, which results in endolysosomal dysfunction. This appears to be particularly relevant in neurons with highly specialized neurites such as cortical motoneurons and Purkinje cells., Author Summary Hereditary spastic paraplegias (HSPs) are inherited disorders characterized by progressive weakness and spasticity of the legs. In HSP patients, nerve fibers connecting cortical motoneurons with spinal cord neurons are progressively lost. HSP subtype 15 (SPG15) is caused by mutations in ZFYVE26, and is characterized by additional cerebellar symptoms. We show that the Zfyve26 protein is broadly expressed in the brain. At the subcellular level Zfyve26 localizes to an intracellular compartment in the endocytic pathway from the plasma membrane to lysosomes, which is part of the degradative system of the cell. Closely resembling the human disease, mice deficient for Zfyve26 develop a progressive spastic gait disorder with cerebellar symptoms and degeneration of both neurons of the motor cortex and Purkinje cells in the cerebellum. Importantly, this degeneration is characterized by the intracellular accumulation of abnormal deposits, which stain positive for the lysosomal marker Lamp1. As Zfyve26 has been shown to interact with the newly identified adaptor complex AP5, which is supposed to be involved in cargo trafficking in the endolysosomal compartment, endolysosomal dysfunction may be caused by a targeting defect upon disruption of Zfyve26. As highly specialized neurons like cortical motoneurons and cerebellar Purkinje cells degenerate, these neurons appear to be particularly dependent on proper endolysosomal function.
- 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.