Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nature ; 613(7942): 179-186, 2023 01.
Article in English | MEDLINE | ID: mdl-36517594

ABSTRACT

Diffuse gliomas, particularly glioblastomas, are incurable brain tumours1. They are characterized by networks of interconnected brain tumour cells that communicate via Ca2+ transients2-6. However, the networks' architecture and communication strategy and how these influence tumour biology remain unknown. Here we describe how glioblastoma cell networks include a small, plastic population of highly active glioblastoma cells that display rhythmic Ca2+ oscillations and are particularly connected to others. Their autonomous periodic Ca2+ transients preceded Ca2+ transients of other network-connected cells, activating the frequency-dependent MAPK and NF-κB pathways. Mathematical network analysis revealed that glioblastoma network topology follows scale-free and small-world properties, with periodic tumour cells frequently located in network hubs. This network design enabled resistance against random damage but was vulnerable to losing its key hubs. Targeting of autonomous rhythmic activity by selective physical ablation of periodic tumour cells or by genetic or pharmacological interference with the potassium channel KCa3.1 (also known as IK1, SK4 or KCNN4) strongly compromised global network communication. This led to a marked reduction of tumour cell viability within the entire network, reduced tumour growth in mice and extended animal survival. The dependency of glioblastoma networks on periodic Ca2+ activity generates a vulnerability7 that can be exploited for the development of novel therapies, such as with KCa3.1-inhibiting drugs.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Mice , Brain/metabolism , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , NF-kappa B/metabolism , MAP Kinase Signaling System , Calcium Signaling , Cell Death , Survival Analysis , Calcium/metabolism
2.
Acta Neuropathol ; 134(3): 507-516, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28401334

ABSTRACT

Pediatric glioblastoma (pedGBM) is an extremely aggressive pediatric brain tumor, accounting for ~6% of all central nervous system neoplasms in children. Approximately half of pedGBM harbor recurrent somatic mutations in histone 3 variants or, infrequently, IDH1/2. The remaining subset of pedGBM is highly heterogeneous, and displays a variety of genomic and epigenetic features. In the current study, we aimed to further stratify an H3-/IDH-wild type (wt) pedGBM cohort assessed through genome-wide molecular profiling. As a result, we identified three molecular subtypes of these tumors, differing in their genomic and epigenetic signatures as well as in their clinical behavior. We designated these subtypes 'pedGBM_MYCN' (enriched for MYCN amplification), 'pedGBM_RTK1' (enriched for PDGFRA amplification) and 'pedGBM_RTK2' (enriched for EGFR amplification). These molecular subtypes were associated with significantly different outcomes, i.e. pedGBM_RTK2 tumors show a significantly longer survival time (median OS 44 months), pedGBM_MYCN display extremely poor outcomes (median OS 14 months), and pedGBM_RTK1 tumors harbor an intermediate prognosis. In addition, the various molecular subtypes of H3-/IDH-wt pedGBM were clearly distinguishable from their adult counterparts, underlining their biological distinctiveness. In conclusion, our study demonstrates significant molecular heterogeneity of H3-/IDH-wt pedGBM in terms of DNA methylation and cytogenetic alterations. The recognition of three molecular subtypes of H3-/IDH-wt pedGBM further revealed close correlations with biological parameters and clinical outcomes and may therefore, be predictive of response to standard treatment protocols, but could also be useful for stratification for novel, molecularly based therapies.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Histones/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Adolescent , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis/genetics , Child , Child, Preschool , DNA Methylation , DNA Mutational Analysis , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Histones/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Male , Prognosis , Promoter Regions, Genetic
3.
Blood ; 120(12): e28-34, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22859608

ABSTRACT

Langerhans cell histiocytosis (LCH) is a clinically and histologically heterogeneous disorder. Its classification as either reactive inflammatory or neoplastic has been a matter of debate. However, the recent finding of frequent BRAFV600E mutations in LCH argues for the latter. The exact cell type that harbors the mutation and is responsible for proliferation remains to be identified. We here apply a BRAFV600E mutation-specific antibody to detect the BRAF mutant cells in lesions from 89 patients with LCH. We found BRAFV600E mutations in 34 of 89 (38%) lesions. In lesions with the BRAFV600E mutation, the majority of cells coexpressing S-100 and CD1a harbored mutant BRAFV600E protein. These cells also expressed CD14 and CD36, whereas various fractions exhibited CD207. On the other hand, CD80 and CD86 expression was also present on BRAFV600E-positive cells. Thus, cells of variable maturation, exhibiting an immunohistochemical profile compatible either with myeloid cell or with dedifferentiated Langerhans cell antigens, carry the BRAFV600E mutation. In conclusion, we identify and characterize the neoplastic cells in LCH with BRAFV600E mutations by applying a mutation-specific marker and demonstrate feasibility for routine screening.


Subject(s)
Biomarkers/metabolism , Histiocytosis, Langerhans-Cell/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Proto-Oncogene Proteins B-raf/metabolism , Blotting, Western , Cells, Cultured , DNA/genetics , Fluorescent Antibody Technique , Histiocytosis, Langerhans-Cell/genetics , Humans , Immunoenzyme Techniques , Mutant Proteins/genetics , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics
4.
Acta Neuropathol ; 127(4): 565-72, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464231

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNST) derive from the Schwann cell or perineurial cell lineage and occur either sporadically or in association with the tumor syndrome neurofibromatosis type 1 (NF1). MPNST often pose a diagnostic challenge due to their frequent lack of pathognomonic morphological or immunohistochemical features. Mutations in the NF1 tumor suppressor gene are found in all NF1-associated and many sporadic MPNST. The presence of NF1 mutation may have the potential to differentiate MPNST from several morphologically similar neoplasms; however, mutation detection is hampered by the size of the gene and the lack of mutational hot spots. Here we describe a newly developed monoclonal antibody binding to the C-terminus of neurofibromin (clone NFC) which was selected for optimal performance in routinely processed formalin-fixed and paraffin-embedded tissue. NFC immunohistochemistry revealed loss of neurofibromin in 22/25 (88 %) of NF1-associated and 26/61 (43 %) of sporadic MPNST. There was a strong association of neurofibromin loss with deletions affecting the NF1 gene (P < 0.01). In a series of 256 soft tissue tumors of different histotypes NFC staining showed loss of neurofibromin in 2/8 myxofibrosarcomas, 2/12 (16 %) pleomorphic liposarcomas, 1/16 (6 %) leiomyosarcomas, and 4/28 (14 %) unclassified undifferentiated pleomorphic sarcomas. However, loss of neurofibromin was not observed in 22 synovial sarcomas, 27 schwannomas, 23 solitary fibrous tumors, 14 low-grade fibromyxoid sarcomas, 50 dedifferentiated liposarcomas, 27 myxoid liposarcomas, 13 angiosarcomas, 9 extraskeletal myxoid chondrosarcomas, and 7 epitheloid sarcomas. Immunohistochemistry using antibody NFC may substantially facilitate sarcoma research and diagnostics.


Subject(s)
Antibodies , Nerve Sheath Neoplasms/diagnosis , Neurilemmoma/diagnosis , Neurilemmoma/metabolism , Neurofibromin 1/immunology , Animals , Cell Line, Transformed , Cloning, Molecular , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Neurofibromin 1/genetics , Schwann Cells/metabolism , Schwann Cells/pathology , Transfection
5.
Sci Immunol ; 9(95): eadj7970, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701193

ABSTRACT

Understanding the mechanisms that regulate T cell immunity is critical for the development of effective therapies for diseases associated with T cell dysfunction, including autoimmune diseases, chronic infections, and cancer. Co-inhibitory "checkpoint molecules," such as programmed cell death protein-1, balance excessive or prolonged immune activation by T cell-intrinsic signaling. Here, by screening for mediators of natural killer (NK) cell recognition on T cells, we identified the immunoglobulin superfamily ligand B7H6 to be highly expressed by activated T cells, including patient-infused CD19-targeting chimeric antigen receptor (CAR) T cells. Unlike other checkpoint molecules, B7H6 mediated NKp30-dependent recognition and subsequent cytolysis of activated T cells by NK cells. B7H6+ T cells were prevalent in the tissue and blood of several diseases, and their abundance in tumor tissue positively correlated with clinical response in a cohort of patients with immune checkpoint inhibitor-treated esophageal cancer. In humanized mouse models, NK cell surveillance via B7H6 limited the persistence and antitumor activity of CAR T cells, and its genetic deletion enhanced T cell proliferation and persistence. Together, we provide evidence of B7H6 protein expression by activated T cells and suggest the B7H6-NKp30 axis as a therapeutically actionable NK cell-dependent immune checkpoint that regulates human T cell function.


Subject(s)
B7 Antigens , Killer Cells, Natural , T-Lymphocytes , Humans , Killer Cells, Natural/immunology , Animals , Mice , B7 Antigens/immunology , T-Lymphocytes/immunology , Natural Cytotoxicity Triggering Receptor 3/immunology , Lymphocyte Activation/immunology , Female , Esophageal Neoplasms/immunology
6.
Acta Neuropathol ; 125(5): 651-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23575898

ABSTRACT

Non-central nervous system hemangiopericytoma (HPC) and solitary fibrous tumor (SFT) are considered by pathologists as two variants of a single tumor entity now subsumed under the entity SFT. Recent detection of frequent NAB2-STAT6 fusions in both, HPC and SFT, provided additional support for this view. On the other hand, current neuropathological practice still distinguishes between HPC and SFT. The present study set out to identify genes involved in the formation of meningeal HPC. We performed exome sequencing and detected the NAB2-STAT6 fusion in DNA of 8/10 meningeal HPC thereby providing evidence of close relationship of these tumors with peripheral SFT. Due to the considerable effort required for exome sequencing, we sought to explore surrogate markers for the NAB2-STAT6 fusion protein. We adopted the Duolink proximity ligation assay and demonstrated the presence of NAB2-STAT6 fusion protein in 17/17 HPC and the absence in 15/15 meningiomas. More practical, presence of the NAB2-STAT6 fusion protein resulted in a strong nuclear signal in STAT6 immunohistochemistry. The nuclear reallocation of STAT6 was detected in 35/37 meningeal HPC and 25/25 meningeal SFT but not in 87 meningiomas representing the most important differential diagnosis. Tissues not harboring the NAB2-STAT6 fusion protein presented with nuclear expression of NAB2 and cytoplasmic expression of STAT6 proteins. In conclusion, we provide strong evidence for meningeal HPC and SFT to constitute variants of a single entity which is defined by NAB2-STAT6 fusion. In addition, we demonstrate that this fusion can be rapidly detected by STAT6 immunohistochemistry which shows a consistent nuclear reallocation. This immunohistochemical assay may prove valuable for the differentiation of HPC and SFT from other mesenchymal neoplasms.


Subject(s)
Hemangiopericytoma/genetics , Meningeal Neoplasms/genetics , Oncogene Proteins, Fusion/physiology , Repressor Proteins/physiology , STAT6 Transcription Factor/physiology , Solitary Fibrous Tumors/genetics , Cohort Studies , Diagnosis, Differential , Exome , Hemangiopericytoma/diagnosis , Hemangiopericytoma/metabolism , Humans , Meningeal Neoplasms/diagnosis , Meningeal Neoplasms/metabolism , RNA, Messenger/metabolism , Solitary Fibrous Tumors/diagnosis , Solitary Fibrous Tumors/metabolism
7.
Acta Neuropathol ; 126(5): 757-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24096618

ABSTRACT

The activating E17K mutation in the AKT1 gene has been detected in several tumor entities. Currently several clinical studies with specific AKT1 inhibitors are under way. To determine whether AKT1 mutations are involved in human tumors of the nervous system, we examined a series of 1,437 tumors including 391 primary intracranial brain tumors and 1,046 tumors of the coverings of the central and peripheral nervous system. AKT1E17K mutations were exclusively seen in meningiomas and occurred in 65 of 958 of these tumors. A strong preponderance was seen in the variant of meningothelial meningioma WHO grade I of basal and spinal localization. In contrast, AKT1E17K mutations were rare in WHO grade II and absent in WHO grade III meningiomas. In order to more effectively detect this mutation, we tested for immunohistochemical markers associated with this alteration. We observed strong up-regulation of SFRP1 expression in all meningiomas with AKT1E17K mutation and in HEK293 cells after transfection with mutant AKT1E17K, but not in meningiomas and HEK293 cells lacking this mutation.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Meningeal Neoplasms/genetics , Meningioma/genetics , Mutation , Proto-Oncogene Proteins c-akt/genetics , Biomarkers, Tumor/analysis , DNA Mutational Analysis , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningioma/metabolism , Meningioma/pathology , Neoplasm Grading , Reverse Transcriptase Polymerase Chain Reaction , Transfection
8.
Acta Neuropathol ; 123(2): 223-33, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22012135

ABSTRACT

Brain metastases (BM) are frequent and carry a dismal prognosis. BRAF V600E mutations are found in a broad range of tumor types and specific inhibitors targeting BRAF V600E protein exist. We analyzed tumoral BRAF V600E-mutant protein expression using the novel mutation-specific antibody VE1 in a series of 1,120 tumor specimens (885 BM, 157 primary tumors, 78 extra-cranial metastases) of 874 BM patients. In 85 cases, we performed validation of immunohistochemical results by BRAF exon 15 gene sequencing. BRAF V600E protein was expressed in BM of 42/76 (55.3%) melanomas, 1/15 (6.7%) ovarian cancers, 4/72 (5.5%) colorectal cancers, 1/355 (0.3%) lung cancers, 2/6 thyroid cancers and 1/2 choriocarcinomas. BRAF V600E expression showed high intra-tumoral homogeneity and was similar in different tumor manifestations of individual patients. VE1 immunohistochemistry and BRAF exon 15 sequencing were congruent in 68/70 (97.1%) cases, but VE1 immunostaining identified small BRAF V600E expressing tumor cell aggregates in 10 cases with inconclusive genetic results. Melanoma patients with BRAF V600E mutant protein expressing tumors were significantly younger at diagnosis of the primary tumor and at operation of BM than patients with non-mutated tumors. In conclusion, expression of BRAF V600E mutant protein occurs in approximately 6% of BM and is consistent in different tumor manifestations of the same patient. Thus, BRAF V600E inhibiting therapies seem feasible in selected BM patients. Immunohistochemical visualization of V600E-mutant BRAF protein is a promising tool for patient stratification. An integrated approach combining both, VE1 immunohistochemistry and genetic analysis may increase the diagnostic accuracy of BRAF mutation analysis.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Genetic Predisposition to Disease/genetics , Point Mutation , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/pathology , Female , Glutamic Acid/genetics , HEK293 Cells , Humans , Immunohistochemistry/methods , Male , Middle Aged , Point Mutation/genetics , Point Mutation/immunology , Proto-Oncogene Proteins B-raf/biosynthesis , Proto-Oncogene Proteins B-raf/immunology , Valine/genetics , Young Adult
9.
Oncogenesis ; 11(1): 57, 2022 Sep 17.
Article in English | MEDLINE | ID: mdl-36115843

ABSTRACT

Glioblastoma is a highly aggressive brain tumor for which there is no cure. The metabolic enzyme 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 4 (PFKFB4) is essential for glioblastoma stem-like cell (GSC) survival but its mode of action is unclear. Understanding the role of PFKFB4 in tumor cell survival could allow it to be leveraged in a cancer therapy. Here, we show the importance of PFKFB4 for glioblastoma growth in vivo in an orthotopic patient derived mouse model. In an evaluation of patient tumor samples of different cancer entities, PFKFB4 protein was found to be overexpressed in prostate, lung, colon, mammary and squamous cell carcinoma, with expression level correlating with tumor grade. Gene expression profiling in PFKFB4-silenced GSCs revealed a downregulation of hypoxia related genes and Western blot analysis confirmed a dramatic reduction of HIF (hypoxia inducible factor) protein levels. Through mass spectrometric analysis of immunoprecipitated PFKFB4, we identified the ubiquitin E3 ligase, F-box only protein 28 (FBXO28), as a new interaction partner of PFKFB4. We show that PFKFB4 regulates the ubiquitylation and subsequent proteasomal degradation of HIF-1α, which is mediated by the ubiquitin ligase activity of FBXO28. This newly discovered function of PFKFB4, coupled with its cancer specificity, provides a new strategy for inhibiting HIF-1α in cancer cells.

10.
Acta Neuropathol ; 122(1): 11-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21638088

ABSTRACT

Activating mutations of the serine threonine kinase v-RAF murine sarcoma viral oncogene homolog B1 (BRAF) are frequent in benign and malignant human tumors and are emerging as an important biomarker. Over 95% of BRAF mutations are of the V600E type and specific small molecular inhibitors are currently under pre-clinical or clinical investigation. BRAF mutation status is determined by DNA-based methods, most commonly by sequencing. Here we describe the development of a monoclonal BRAF V600E mutation-specific antibody that can differentiate BRAF V600E and wild type protein in routinely processed formalin-fixed and paraffin-embedded tissue. A total of 47 intracerebral melanoma metastases and 21 primary papillary thyroid carcinomas were evaluated by direct sequencing of BRAF and by immunohistochemistry using the BRAF V600E mutation-specific antibody clone VE1. Correlation of VE1 immunohistochemistry and BRAF sequencing revealed a perfect match for both papillary thyroid carcinomas and melanoma metastases. The staining intensity in BRAF V600E mutated tumor samples ranged from weak to strong. The generally homogenous VE1 staining patterns argue against a clonal heterogeneity of the tumors investigated. Caution is essential when only poorly preserved tissue is available for VE1 immunohistochemical analysis or when tissues with only little total BRAF protein are analyzed. Immunohistochemistry using antibody VE1 may substantially facilitate molecular analysis of BRAF V600E status for diagnostic, prognostic, and predictive purposes.


Subject(s)
Antibodies, Monoclonal , Biomarkers, Tumor/genetics , Immunohistochemistry/methods , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Animals , Antibodies, Monoclonal/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Carcinoma, Papillary/diagnosis , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Female , Humans , Melanoma/diagnosis , Melanoma/metabolism , Melanoma/secondary , Mice , Mice, Inbred C57BL , Predictive Value of Tests , Prognosis , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
11.
Neurobiol Dis ; 33(2): 243-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19027068

ABSTRACT

17-AAG is a selective HSP90-inhibitor that exhibited therapeutic activity in cancer. In this study three glioblastoma cell lines (U87, LN229 and U251) were treated with 17-AAG, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or the combination of both. Treatment with subtoxic doses of 17-AAG in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces rapid apoptosis in TRAIL-resistant glioma cells, suggesting that this combined treatment may offer an attractive strategy for treating gliomas. 17-AAG treatment down-regulated survivin through proteasomal degradation. In addition, over-expression of survivin attenuated cytotoxicity induced by the combination of 17-AAG and TRAIL. In summary, survivin is a key regulator of TRAIL-17-AAG mediated cell death in malignant glioma.


Subject(s)
Apoptosis/drug effects , Benzoquinones/pharmacology , Glioblastoma/pathology , Lactams, Macrocyclic/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Benzoquinones/administration & dosage , Benzoquinones/therapeutic use , Blotting, Western , Brain/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytotoxins/therapeutic use , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Flow Cytometry , Glioblastoma/drug therapy , Glioblastoma/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/therapeutic use , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Survivin , TNF-Related Apoptosis-Inducing Ligand/therapeutic use
12.
Neurobiol Dis ; 34(2): 259-66, 2009 May.
Article in English | MEDLINE | ID: mdl-19385057

ABSTRACT

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising cancer therapeutic. The main obstacle in TRAIL-based therapy is that many glioma cells are resistant. In this study glioblastoma cell lines, human glioblastoma short-term cultures and human astrocytes were treated with 3-keto-N-aminoethylaminoethylcaproyldihydrocinnamoyl cyclopamine (KAAD-cyclopamine), tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) or the combination of both. Single treatment with KAAD-cyclopamine or TRAIL does not induce cytotoxicity in malignant glioma cells. However, treatment with KAAD-cyclopamine in combination with TRAIL induces rapid apoptosis in TRAIL-resistant glioma cells. Notably, normal human astrocytes were not affected by the combination treatment consisting of KAAD-cyclopamine and TRAIL. KAAD-cyclopamine led to an upregulation of death receptor 4 and 5 and down-regulation of bcl-2 and c-FLIP. Furthermore, overexpression of both bcl-2 and c-FLIP attenuated KAAD-cyclopamine facilitated TRAIL-mediated apoptosis. Taken together,we provided evidence that KAAD-cyclopamine facilitated TRAIL-mediated apoptosis at the level of the intrinsic and extrinsic apoptotic pathways in malignant glioma cells.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cinnamates/pharmacology , Glioma/drug therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Veratrum Alkaloids/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/physiology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/physiopathology , CASP8 and FADD-Like Apoptosis Regulating Protein/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Cinnamates/therapeutic use , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Glioma/metabolism , Glioma/physiopathology , Humans , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Up-Regulation/drug effects , Up-Regulation/physiology , Veratrum Alkaloids/therapeutic use
13.
Neuro Oncol ; 11(2): 122-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18971417

ABSTRACT

The flavonoid quercetin has been reported to inhibit the proliferation of cancer cells, whereas it has no effect on nonneoplastic cells. U87-MG, U251, A172, LN229, and U373 malignant glioma cells were treated with quercetin (50-200 microM). Quercetin did not cause cytotoxicity 24 h after treatment. Combining quercetin with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) strongly augmented TRAIL-mediated apoptosis in U87-MG, U251, A172, and LN229 glioma cells; U373 cells could not be sensitized by quercetin to TRAIL-mediated apoptosis. TRAIL-induced apoptosis was enhanced by quercetin-induced reduction of survivin protein levels. Upon treatment with quercetin, the protein level of survivin was strongly suppressed in U87-MG, U251, and A172 but not in U373 glioma cells. Quercetin exposure resulted in proteasomal degradation of survivin. TRAIL-quercetin-induced apoptosis was markedly reduced by overexpression of survivin. In addition, upon treatment with quercetin, downregulation of survivin was also regulated by the Akt pathway. Taken together, the results of the present study suggest that quercetin sensitizes glioma cells to death-receptor-mediated apoptosis by suppression of inhibitor of the apoptosis protein survivin.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Glioma/pathology , Microtubule-Associated Proteins/metabolism , Quercetin/pharmacology , Receptors, Death Domain/metabolism , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Glioma/drug therapy , Glioma/metabolism , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Death Domain/genetics , Survivin , TNF-Related Apoptosis-Inducing Ligand/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
14.
Acta Neuropathol ; 117(4): 445-56, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19214542

ABSTRACT

Tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL/Apo2L) is a promising cancer drug. However, many tumours are resistant to TRAIL-based therapies. Glioma cells with stem cell features (SCG), such as CD133 expression and neurosphere formation, have been recently identified to be more resistant to cytotoxic drugs than glioma cells lacking stem-cell-like features (NSCGs). Here we report that SCGs are completely resistant to 100-2,000 ng/ml TRAIL, whereas NSCGs revealed a moderate sensitivity to TRAIL. We found that SCGs exhibited only low levels of caspase-8 mRNA and protein, known to be indispensable for TRAIL-induced apoptosis. In addition, we detected hypermethylation of CASP8 promoter in SCGs, whereas NSCGs exhibited a non-methylated CASP8 promoter. Reexpression of caspase-8 by 5-Aza-2'-deoxycytidine was not sufficient to restore TRAIL sensitivity in SCGs cells, suggesting that additional factors cause TRAIL resistance in SCGs. Our data suggest that therapy with TRAIL, either as monotherapy or in combination with demethylating agents, is not effective in treating glioblastoma because SCGs are not targeted by such treatment.


Subject(s)
Caspase 8/genetics , Drug Resistance, Neoplasm , Glioma/pathology , Promoter Regions, Genetic , Stem Cells/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , AC133 Antigen , Antigens, CD/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 8/metabolism , Cell Death/drug effects , Cycloheximide/pharmacology , DNA Methylation , Decitabine , Down-Regulation , Glioma/metabolism , Glycoproteins/metabolism , Humans , Peptides/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Tumor Cells, Cultured
15.
Neurosci Lett ; 453(2): 92-7, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19356600

ABSTRACT

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising cancer drug. One obstacle in TRAIL-based therapies is that many cancer cells, including gliomas, are resistant towards TRAIL. In this study one glioblastoma cell line, one human short-term glioblastoma culture and human astrocytes were treated with genistein, tumour necrosis factor-related apoptosis-inducing ligand or the combination of both. Single treatment with genistein or TRAIL does not induce cytotoxicity in malignant glioma cells. However, treatment with genistein in combination with TRAIL induces rapid apoptosis in TRAIL-resistant glioma cells. Notably, normal human astrocytes were not affected by the combination treatment consisting of genistein and TRAIL. Genistein enhanced proteasomal degradation of the short isoform of c-FLIP. Importantly, over-expression of only the short isoform of c-FLIP attenuated genistein TRAIL-mediated cytotoxicity. Taken together, we gave evidence that genistein facilitated TRAIL-mediated apoptosis at the level of the extrinsic apoptotic pathways in malignant glioma cells.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Astrocytes/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Genistein/pharmacology , Glioblastoma/drug therapy , Proteasome Endopeptidase Complex/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Astrocytes/metabolism , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein/classification , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Humans , Tumor Cells, Cultured
16.
Mol Cancer Ther ; 7(11): 3566-74, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19001439

ABSTRACT

Resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) limits its potential as a drug for cancer therapy. Here, we report that kaempferol, a bioactive plant flavonoid, sensitizes U251 and U87 glioma cells to TRAIL-mediated apoptosis. In contrast, U373 cells are not affected by kaempferol treatment. Treatment of kaempferol alone for 24 h did not induce apoptosis in the cell lines. We provide evidence that TRAIL-induced apoptosis is partially driven by kaempferol-mediated reduction of survivin protein levels. On kaempferol treatment, proteasomal degradation of survivin was observed. Inhibition of proteasomal degradation with MG132 in kaempferol-treated cells restored survivin protein levels in both glial cell lines. Consequently, overexpression of survivin attenuated TRAIL-kaempferol-induced apoptosis. In addition, we show that kaempferol mediates down-regulation of phosphorylated Akt, thereby further reducing survivin protein level. Furthermore, the blockage of the serine/threonine kinase Akt activity by kaempferol is important for inhibition of survivin because active phosphorylated Akt enhances the stability of survivin. However, we also show that the combined treatment of TRAIL and kaempferol induces cleavage (activation) of caspase-8, thereby exerting a proapoptotic effect independent of survivin known not to inhibit caspase-8 activation. Other effects induced by kaempferol were suppression of X-linked inhibitor of apoptosis proteins as the antiapoptotic members of the Bcl-2 family, Bcl-2, Bcl-xL, and Mcl-1 in a concentration-dependent manner. In summary, we showed that suppression of survivin is an essential mechanism in TRAIL-kaempferol-mediated apoptosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Glioma/metabolism , Kaempferols/pharmacology , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Caspases/metabolism , Cell Line, Tumor , Glioma/drug therapy , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , Survivin
17.
Neuro Oncol ; 21(2): 214-221, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30252101

ABSTRACT

BACKGROUND: Wingless-activated medulloblastoma (WNT MB) represents a well-characterized molecular variant accounting for 10-15% of all MB and is associated with a favorable clinical outcome. Patients with localized WNT MBs could benefit from de-intensification of combined treatment, which would require an accurate diagnosis of these tumors. However, despite the presence of molecular features related with a WNT MB signature (nuclear ß-catenin immunoexpression, CTNNB1 mutation, and monosomy 6), a prompt and reliable diagnostic verification of these tumors is not yet feasible. METHODS: In the current study, we analyzed 78 samples of WNT MB treated in a single institute through genome-wide DNA methylation and targeted next generation sequencing to elaborate an optimal method for WNT MB molecular verification. RESULTS: We found that DNA methylation profiling discloses significant advantages for molecular diagnostic of WNT MB. All other "routine" methods applied, such as ß-catenin immunohistochemistry, CTNNB1 mutation analysis, and detection of monosomy 6, failed to identify all WNT MB cases. Survival analysis revealed that application of a reduced radiotherapy protocol for WNT MB treatment had no influence on patients' survival. Only one patient died due to local relapse but recurrent tumor was pathologically and molecularly diagnosed as a secondary glioblastoma. CONCLUSIONS: DNA methylation analysis should be considered as a method of choice for further clinically relevant stratification of WNT MB and for correct diagnosis of the recurrent tumors. WNT MB patients with localized disease could benefit from treatment de-intensification.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Neoplasms/diagnosis , DNA Methylation , Gene Expression Regulation, Neoplastic , Medulloblastoma/diagnosis , Wnt Proteins/metabolism , Adolescent , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Male , Medulloblastoma/genetics , Medulloblastoma/metabolism , Prognosis , Retrospective Studies
18.
Brain Pathol ; 28(5): 656-662, 2018 09.
Article in English | MEDLINE | ID: mdl-28990704

ABSTRACT

Epithelioid glioblastoma (eGBM) is a newly defined and rare GBM variant in the current WHO 2016 classification. BRAF V600E mutation is overrepresented in these tumors and there is known some morphological overlap with anaplastic epithelioid PXA (ePXA). In order to further elucidate this diagnostic category, we molecularly characterized 64 pediatric and adult examples initially diagnosed as "eGBM." Tumors were analyzed using array based methylation and direct sequencing of the BRAF and TERT genes. Our results demonstrated considerable molecular and clinical heterogeneity among eGBM cohort. Methylation patterns, copy number alterations, and mutational analysis data, in combination with clinical findings disclosed three different, well established tumor subtypes: (i) PXA-like tumors with favorable prognosis, predominantly in children and young adults (38), (ii) IDHwt GBM-like tumors with poor prognosis, mainly occurring in older adults, albeit with more frequent BRAF mutations (17), and (iii) RTK1 pediatric GBM-like neoplasms of intermediate prognosis in children and young adults, associated with chromothripsis and frequent PDGFRA amplifications (9). We conclude that the histopathologically defined eGBM do not represent a single diagnostic entity, but rather at least three molecularly and biologically distinct categories. Therefore, additional molecular testing through genome-wide molecular profiling is recommended to further stratify these rare cases.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Child , Child, Preschool , Cohort Studies , Computational Biology , DNA Copy Number Variations , Epigenesis, Genetic , Female , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Male , Middle Aged , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Young Adult
19.
Nat Med ; 24(8): 1192-1203, 2018 08.
Article in English | MEDLINE | ID: mdl-29988124

ABSTRACT

The oncometabolite (R)-2-hydroxyglutarate (R-2-HG) produced by isocitrate dehydrogenase (IDH) mutations promotes gliomagenesis via DNA and histone methylation. Here, we identify an additional activity of R-2-HG: tumor cell-derived R-2-HG is taken up by T cells where it induces a perturbation of nuclear factor of activated T cells transcriptional activity and polyamine biosynthesis, resulting in suppression of T cell activity. IDH1-mutant gliomas display reduced T cell abundance and altered calcium signaling. Antitumor immunity to experimental syngeneic IDH1-mutant tumors induced by IDH1-specific vaccine or checkpoint inhibition is improved by inhibition of the neomorphic enzymatic function of mutant IDH1. These data attribute a novel, non-tumor cell-autonomous role to an oncometabolite in shaping the tumor immune microenvironment.


Subject(s)
Glutarates/metabolism , Immunity , T-Lymphocytes/immunology , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation , Glioma/genetics , Glioma/immunology , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mutation/genetics , NFATC Transcription Factors/metabolism , Paracrine Communication , Polyamines/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
20.
Arch Neurol ; 69(4): 523-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22158715

ABSTRACT

OBJECTIVE: To analyze infiltration patterns of IDH1 mutant diffuse gliomas into the brain by identification of single tumor cells applying an antibody specific to mutant IDH1 R132H protein. DESIGN: Immunohistochemical analysis. SETTING: University hospital. PATIENTS: Whole-brain and hemisphere sections of 4 patients diagnosed with diffuse glioma. RESULTS: Tumor cells were identified in areas that appeared inconspicuous macroscopically and at histological analysis with respect to cellularity, cellular pleomorphism, or mitotic activity in all cases. CONCLUSION: Detection of single tumor cells throughout the brain demonstrates diffuse glioma to represent systemic brain disease.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Adult , Aged , Brain/metabolism , Brain/pathology , Functional Laterality , Humans , Magnetic Resonance Imaging , Male , Single-Cell Analysis/methods
SELECTION OF CITATIONS
SEARCH DETAIL