Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nature ; 606(7916): 945-952, 2022 06.
Article in English | MEDLINE | ID: mdl-35732742

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a heterogenous neurodegenerative disorder that affects motor neurons and voluntary muscle control1. ALS heterogeneity includes the age of manifestation, the rate of progression and the anatomical sites of symptom onset. Disease-causing mutations in specific genes have been identified and define different subtypes of ALS1. Although several ALS-associated genes have been shown to affect immune functions2, whether specific immune features account for ALS heterogeneity is poorly understood. Amyotrophic lateral sclerosis-4 (ALS4) is characterized by juvenile onset and slow progression3. Patients with ALS4 show motor difficulties by the time that they are in their thirties, and most of them require devices to assist with walking by their fifties. ALS4 is caused by mutations in the senataxin gene (SETX). Here, using Setx knock-in mice that carry the ALS4-causative L389S mutation, we describe an immunological signature that consists of clonally expanded, terminally differentiated effector memory (TEMRA) CD8 T cells in the central nervous system and the blood of knock-in mice. Increased frequencies of antigen-specific CD8 T cells in knock-in mice mirror the progression of motor neuron disease and correlate with anti-glioma immunity. Furthermore, bone marrow transplantation experiments indicate that the immune system has a key role in ALS4 neurodegeneration. In patients with ALS4, clonally expanded TEMRA CD8 T cells circulate in the peripheral blood. Our results provide evidence of an antigen-specific CD8 T cell response in ALS4, which could be used to unravel disease mechanisms and as a potential biomarker of disease state.


Subject(s)
Amyotrophic Lateral Sclerosis , CD8-Positive T-Lymphocytes , Clone Cells , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Clone Cells/pathology , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Knock-In Techniques , Mice , Motor Neurons/pathology , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutation , RNA Helicases/genetics , RNA Helicases/metabolism
3.
Hum Mol Genet ; 29(R2): R226-R235, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32639003

ABSTRACT

The last decade's progress unraveling the mutational landscape of all age groups of cancer has uncovered mutations in histones as vital contributors of tumorigenesis. Here we review three new aspects of oncogenic histones: first, the identification of additional histone mutations potentially contributing to cancer formation; second, tumors expressing histone mutations to study the crosstalk of post-translational modifications, and; third, development of sophisticated biological model systems to reproduce tumorigenesis. At the outset, we recapitulate the firstly discovered histone mutations in pediatric and adolescent tumors of the brain and bone, which still remain the most pronounced histone alterations in cancer. We branch out to discuss the ramifications of histone mutations, including novel ones, that stem from altered protein-protein interactions of cognate histone modifiers as well as the stability of the nucleosome. We close by discussing animal models of oncogenic histones that reproduce tumor formation molecularly and morphologically and the prospect of utilizing them for drug testing, leading to efficient treatment and cure of deadly cancers with histone mutations.


Subject(s)
Disease Models, Animal , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/genetics , Mutation , Neoplasms/genetics , Nucleosomes/genetics , Animals , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice
4.
Genes Dev ; 24(19): 2205-18, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20889717

ABSTRACT

Gene rearrangement in the form of an intragenic deletion is the primary mechanism of oncogenic mutation of the epidermal growth factor receptor (EGFR) gene in gliomas. However, the incidence of platelet-derived growth factor receptor-α (PDGFRA) gene rearrangement in these tumors is unknown. We investigated the PDGFRA locus in PDGFRA-amplified gliomas and identified two rearrangements, including the first case of a gene fusion between kinase insert domain receptor (KDR) (VEGFRII) and the PDGFRA gene, and six cases of PDGFRA(Δ8, 9), an intragenic deletion rearrangement. The PDGFRA(Δ8, 9) mutant was common, being present in 40% of the glioblastoma multiformes (GBMs) with PDGFRA amplification. Tumors with these two types of PDGFRA rearrangement displayed histologic features of oligodendroglioma, and the gene products of both rearrangements showed constitutively elevated tyrosine kinase activity and transforming potential that was reversed by PDGFR blockade. These results suggest the possibility that these PDGFRA mutants behave as oncogenes in this subset of gliomas, and that the prevalence of such rearrangements may have been considerably underestimated.


Subject(s)
Gene Rearrangement , Glioblastoma/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Amino Acid Sequence , Base Sequence , Benzamides , Gene Dosage , Gene Fusion/genetics , Glioblastoma/pathology , Humans , Imatinib Mesylate , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutation/genetics , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Phthalazines/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Transformation, Genetic/drug effects
5.
Genes Dev ; 23(24): 2806-11, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20008931

ABSTRACT

The miR-17 approximately 92 cluster is frequently amplified or overexpressed in human cancers and has emerged as the prototypical oncogenic polycistron microRNA (miRNA). miR-17 approximately 92 is a direct transcriptional target of c-Myc, and experiments in a mouse model of B-cell lymphomas have shown cooperation between these two oncogenes. However, both the molecular mechanism underlying this cooperation and the individual miRNAs that are responsible for it are unknown. By using a conditional knockout allele of miR-17 approximately 92, we show here that sustained expression of endogenous miR-17 approximately 92 is required to suppress apoptosis in Myc-driven B-cell lymphomas. Furthermore, we show that among the six miRNAs that are encoded by miR-17 approximately 92, miR-19a and miR-19b are absolutely required and largely sufficient to recapitulate the oncogenic properties of the entire cluster. Finally, by combining computational target prediction, gene expression profiling, and an in vitro screening strategy, we identify a subset of miR-19 targets that mediate its prosurvival activity.


Subject(s)
Genes, myc/genetics , Lymphoma, B-Cell/genetics , MicroRNAs/genetics , Animals , Cell Line, Tumor , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/metabolism
6.
EMBO J ; 30(19): 4084-97, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21915099

ABSTRACT

In tumours, aberrant splicing generates variants that contribute to multiple aspects of tumour establishment, progression and maintenance. We show that in glioblastoma multiforme (GBM) specimens, death-domain adaptor protein Insuloma-Glucagonoma protein 20 (IG20) is consistently aberrantly spliced to generate an antagonist, anti-apoptotic isoform (MAP-kinase activating death domain protein, MADD), which effectively redirects TNF-α/TRAIL-induced death signalling to promote survival and proliferation instead of triggering apoptosis. Splicing factor hnRNPH, which is upregulated in gliomas, controls this splicing event and similarly mediates switching to a ligand-independent, constitutively active Recepteur d'Origine Nantais (RON) tyrosine kinase receptor variant that promotes migration and invasion. The increased cell death and the reduced invasiveness caused by hnRNPH ablation can be rescued by the targeted downregulation of IG20/MADD exon 16- or RON exon 11-containing variants, respectively, using isoform-specific knockdown or splicing redirection approaches. Thus, hnRNPH activity appears to be involved in the pathogenesis and progression of malignant gliomas as the centre of a splicing oncogenic switch, which might reflect reactivation of stem cell patterns and mediates multiple key aspects of aggressive tumour behaviour, including evasion from apoptosis and invasiveness.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Alternative Splicing , Animals , Cerebral Cortex/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Exons , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells , Humans , MAP Kinase Signaling System , Mice , RNA Splicing , Regulatory Elements, Transcriptional
7.
Cancer Cell ; 10(6): 454-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157785

ABSTRACT

The concepts of stem cells being resistant to therapy, stem-like cells existing in brain tumors, and these tumors initially responding to therapy followed by recurrence are well documented. On this foundation, a recent paper in Nature has demonstrated that CD133-expressing glioma cells in vivo and in culture are relatively resistant to radiation. The mechanism of resistance involves the cell-cycle-regulating proteins CHK1/CHK2. The data raise many questions about the details of radiobiology of stem-like cells in their native environment within tumors in vivo. These answers may lead to better optimization of radiation treatments and schedules for these patients.


Subject(s)
Brain Neoplasms/radiotherapy , Neoplastic Stem Cells/pathology , Radiation Tolerance , AC133 Antigen , Antigens, CD/analysis , Brain Neoplasms/pathology , Glycoproteins/analysis , Humans , Peptides/analysis
8.
Nature ; 448(7157): 1063-7, 2007 Aug 30.
Article in English | MEDLINE | ID: mdl-17728759

ABSTRACT

The acetyl-transferase Tip60 might influence tumorigenesis in multiple ways. First, Tip60 is a co-regulator of transcription factors that either promote or suppress tumorigenesis, such as Myc and p53. Second, Tip60 modulates DNA-damage response (DDR) signalling, and a DDR triggered by oncogenes can counteract tumour progression. Using E(mu)-myc transgenic mice that are heterozygous for a Tip60 gene (Htatip) knockout allele (hereafter denoted as Tip60+/- mice), we show that Tip60 counteracts Myc-induced lymphomagenesis in a haplo-insufficient manner and in a time window that is restricted to a pre- or early-tumoral stage. Tip60 heterozygosity severely impaired the Myc-induced DDR but caused no general DDR defect in B cells. Myc- and p53-dependent transcription were not affected, and neither were Myc-induced proliferation, activation of the ARF-p53 tumour suppressor pathway or the resulting apoptotic response. We found that the human TIP60 gene (HTATIP) is a frequent target for mono-allelic loss in human lymphomas and head-and-neck and mammary carcinomas, with concomitant reduction in mRNA levels. Immunohistochemical analysis also demonstrated loss of nuclear TIP60 staining in mammary carcinomas. These events correlated with disease grade and frequently concurred with mutation of p53. Thus, in both mouse and human, Tip60 has a haplo-insufficient tumour suppressor activity that is independent from-but not contradictory with-its role within the ARF-p53 pathway. We suggest that this is because critical levels of Tip60 are required for mounting an oncogene-induced DDR in incipient tumour cells, the failure of which might synergize with p53 mutation towards tumour progression.


Subject(s)
DNA Damage , Histone Acetyltransferases/metabolism , Oncogene Protein p55(v-myc)/metabolism , Oncogenes/genetics , Tumor Suppressor Proteins/metabolism , Alleles , Animals , B-Lymphocytes/metabolism , Carcinoma/genetics , Carcinoma/pathology , Cells, Cultured , Genes, Tumor Suppressor , Genes, myc/genetics , Heterozygote , Histone Acetyltransferases/genetics , Homeostasis , Humans , Lymphoma/genetics , Lymphoma/pathology , Lysine Acetyltransferase 5 , Mice , Mice, Transgenic , Oncogene Protein p55(v-myc)/genetics , Trans-Activators , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
9.
Acta Neuropathol ; 123(4): 573-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22302101

ABSTRACT

Development of the cerebellum occurs postnatally and is marked by a rapid proliferation of cerebellar granule neuron precursors (CGNPs). CGNPs are the cells of origin for SHH-driven medulloblastoma, the most common malignant brain tumor in children. Here, we investigated the role of ERK, JNK, and p38 mitogen-activated protein kinases in CGNP proliferation. We found high levels of p38α in proliferating CGNPs. Concomitantly, members of the p38 pathway, such as ASK1, MKK3 and ATF-2, were also elevated. Inhibition of the Shh pathway or CGNP proliferation blunts p38α levels, irrespective of Shh treatment. Strikingly, p38α levels were high in vivo in the external granule layer of the postnatal cerebellum, Shh-dependent mouse medulloblastomas and human medulloblastomas of the SHH subtype. Finally, knocking down p38α by short hairpin RNA-carrying lentiviruses as well as the pharmacologically inhibiting of its kinase activity caused a marked decrease in CGNP proliferation, underscoring its requirement for Shh-dependent proliferation in CGNPs. The inhibition of p38α also caused a decrease in Gli1 and N-myc transcript levels, consistent with reduced proliferation. These findings suggest p38 inhibition as a potential way to increase the efficacy of treatments available for malignancies associated with deregulated SHH signaling, such as basal cell carcinoma and medulloblastoma.


Subject(s)
Cell Proliferation/physiology , Cerebellum/enzymology , Neural Stem Cells/enzymology , Neurons/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Brain/enzymology , Cells, Cultured , Gene Knockdown Techniques , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/enzymology , Mice , Mice, Transgenic , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics
10.
JCI Insight ; 7(7)2022 04 08.
Article in English | MEDLINE | ID: mdl-35393952

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors, and patient survival has not changed despite many therapeutic efforts, emphasizing the urgent need for effective treatments. Here, we evaluated the anti-DIPG effect of the oncolytic adenovirus Delta-24-ACT, which was engineered to express the costimulatory ligand 4-1BBL to potentiate the antitumor immune response of the virus. Delta-24-ACT induced the expression of functional 4-1BBL on the membranes of infected DIPG cells, which enhanced the costimulation of CD8+ T lymphocytes. In vivo, Delta-24-ACT treatment of murine DIPG orthotopic tumors significantly improved the survival of treated mice, leading to long-term survivors that developed immunological memory against these tumors. In addition, Delta-24-ACT was safe and caused no local or systemic toxicity. Mechanistic studies showed that Delta-24-ACT modulated the tumor-immune content, not only increasing the number, but also improving the functionality of immune cells. All of these data highlight the safety and potential therapeutic benefit of Delta-24-ACT the treatment of patients with DIPG.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Oncolytic Virotherapy , Adenoviridae , Animals , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/therapy , Humans , Mice
11.
Trends Cell Biol ; 16(9): 433-42, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16904321

ABSTRACT

The Tip60 histone acetyltransferase is part of an evolutionarily conserved multisubunit complex, NuA4, which is recruited by many transcription factors to their target promoters, where it is thought to participate in histone acetylation and transcriptional activation. These transcription factors include tumor promoters and also tumor suppressors, such as p53, which links Tip60 to DNA damage responses. Tip60 also has transcription-independent roles in DNA damage responses. First, independently from NuA4, Tip60 binds the kinases ataxia-telangiectasia mutated (ATM) and DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and participates in their activation by DNA double-strand breaks. Second, NuA4 is recruited to the chromatin surrounding the breaks and, through a series of chromatin modifications, contributes to the dynamics of DNA repair. These molecular activities might endow Tip60 with multiple and potentially antagonistic biological functions.


Subject(s)
DNA Damage , DNA Repair , Histone Acetyltransferases/metabolism , Acetyltransferases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Lysine Acetyltransferase 5 , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Cancer Discov ; 11(3): 754-777, 2021 03.
Article in English | MEDLINE | ID: mdl-33361384

ABSTRACT

Glioblastoma is a lethal brain tumor that exhibits heterogeneity and resistance to therapy. Our understanding of tumor homeostasis is limited by a lack of genetic tools to selectively identify tumor states and fate transitions. Here, we use glioblastoma subtype signatures to construct synthetic genetic tracing cassettes and investigate tumor heterogeneity at cellular and molecular levels, in vitro and in vivo. Through synthetic locus control regions, we demonstrate that proneural glioblastoma is a hardwired identity, whereas mesenchymal glioblastoma is an adaptive and metastable cell state driven by proinflammatory and differentiation cues and DNA damage, but not hypoxia. Importantly, we discovered that innate immune cells divert glioblastoma cells to a proneural-to-mesenchymal transition that confers therapeutic resistance. Our synthetic genetic tracing methodology is simple, scalable, and widely applicable to study homeostasis in development and diseases. In glioblastoma, the method causally links distinct (micro)environmental, genetic, and pharmacologic perturbations and mesenchymal commitment. SIGNIFICANCE: Glioblastoma is heterogeneous and incurable. Here, we designed synthetic reporters to reflect the transcriptional output of tumor cell states and signaling pathways' activity. This method is generally applicable to study homeostasis in normal tissues and diseases. In glioblastoma, synthetic genetic tracing causally connects cellular and molecular heterogeneity to therapeutic responses.This article is highlighted in the In This Issue feature, p. 521.


Subject(s)
Cell Communication , Gene Expression Regulation, Neoplastic , Glioblastoma/etiology , Glioblastoma/pathology , Immunity, Innate , Biomarkers, Tumor , Cell Communication/genetics , Disease Susceptibility , Glioblastoma/metabolism , Humans , Immunity, Innate/genetics , Neoplasm Grading , Neoplasm Staging , Transcriptome , Tumor Microenvironment
13.
Mol Cancer Ther ; 20(6): 1029-1038, 2021 06.
Article in English | MEDLINE | ID: mdl-33846235

ABSTRACT

Glioblastoma (GBM) is the most frequent and aggressive primary tumor type in the central nervous system in adults. Resistance to chemotherapy remains one of the major obstacles in GBM treatment. Identifying and overcoming the mechanisms of therapy resistance is instrumental to develop novel therapeutic approaches for patients with GBM. To determine the major drivers of temozolomide (TMZ) sensitivity, we performed shRNA screenings in GBM lines with different O6-methylguanine-DNA methyl-transferase (MGMT) status. We then evaluated dianhydrogalactitol (Val-083), a small alkylating molecule that induces interstrand DNA crosslinking, as a potential treatment to bypass TMZ-resistance mechanisms. We found that loss of mismatch repair (MMR) components and MGMT expression are mutually exclusive mechanisms driving TMZ resistance in vitro Treatment of established GBM cells and tumorsphere lines with Val-083 induces DNA damage and cell-cycle arrest in G2-M phase, independently of MGMT or MMR status, thus circumventing conventional resistance mechanisms to TMZ. Combination of TMZ and Val-083 shows a synergic cytotoxic effect in tumor cells in vitro, ex vivo, and in vivo We propose this combinatorial treatment as a potential approach for patients with GBM.


Subject(s)
Dianhydrogalactitol/therapeutic use , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , Temozolomide/pharmacology , Animals , Cell Line, Tumor , Dianhydrogalactitol/pharmacology , Humans , Mice , Transfection , Xenograft Model Antitumor Assays
14.
Elife ; 102021 08 17.
Article in English | MEDLINE | ID: mdl-34399888

ABSTRACT

The molecular basis underlying glioblastoma (GBM) heterogeneity and plasticity is not fully understood. Using transcriptomic data of human patient-derived brain tumor stem cell lines (BTSCs), classified based on GBM-intrinsic signatures, we identify the AP-1 transcription factor FOSL1 as a key regulator of the mesenchymal (MES) subtype. We provide a mechanistic basis to the role of the neurofibromatosis type 1 gene (NF1), a negative regulator of the RAS/MAPK pathway, in GBM mesenchymal transformation through the modulation of FOSL1 expression. Depletion of FOSL1 in NF1-mutant human BTSCs and Kras-mutant mouse neural stem cells results in loss of the mesenchymal gene signature and reduction in stem cell properties and in vivo tumorigenic potential. Our data demonstrate that FOSL1 controls GBM plasticity and aggressiveness in response to NF1 alterations.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplastic Stem Cells/pathology , Neurofibromin 1/genetics , Proto-Oncogene Proteins c-fos/genetics , Cell Line, Tumor , Humans , Neurofibromin 1/metabolism , Proto-Oncogene Proteins c-fos/metabolism
15.
Cancer Res ; 81(8): 2101-2115, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33483373

ABSTRACT

The tumor microenvironment plays an essential role in supporting glioma stemness and radioresistance. Following radiotherapy, recurrent gliomas form in an irradiated microenvironment. Here we report that astrocytes, when pre-irradiated, increase stemness and survival of cocultured glioma cells. Tumor-naïve brains increased reactive astrocytes in response to radiation, and mice subjected to radiation prior to implantation of glioma cells developed more aggressive tumors. Extracellular matrix derived from irradiated astrocytes were found to be a major driver of this phenotype and astrocyte-derived transglutaminase 2 (TGM2) was identified as a promoter of glioma stemness and radioresistance. TGM2 levels increased after radiation in vivo and in recurrent human glioma, and TGM2 inhibitors abrogated glioma stemness and survival. These data suggest that irradiation of the brain results in the formation of a tumor-supportive microenvironment. Therapeutic targeting of radiation-induced, astrocyte-derived extracellular matrix proteins may enhance the efficacy of standard-of-care radiotherapy by reducing stemness in glioma. SIGNIFICANCE: These findings presented here indicate that radiotherapy can result in a tumor-supportive microenvironment, the targeting of which may be necessary to overcome tumor cell therapeutic resistance and recurrence. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2101/F1.large.jpg.


Subject(s)
Astrocytes/enzymology , Brain Neoplasms/radiotherapy , Brain/radiation effects , GTP-Binding Proteins/metabolism , Glioblastoma/radiotherapy , Neoplastic Stem Cells , Transglutaminases/metabolism , Tumor Microenvironment/radiation effects , Animals , Astrocytes/radiation effects , Brain/cytology , Brain/physiology , Brain Neoplasms/pathology , Cell Survival/physiology , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix/radiation effects , Female , GTP-Binding Proteins/antagonists & inhibitors , Glioblastoma/pathology , Glioma/pathology , Glioma/radiotherapy , Humans , Male , Mice , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/physiology , Protein Glutamine gamma Glutamyltransferase 2 , Radiation Tolerance , Transglutaminases/antagonists & inhibitors , Tumor Microenvironment/physiology
16.
Cancers (Basel) ; 13(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478100

ABSTRACT

(1) Background: Despite the indisputable effectiveness of dexamethasone (DEXA) to reduce inflammation in glioblastoma (GBM) patients, its influence on tumour progression and radiotherapy response remains controversial. (2) Methods: We analysed patient data and used expression and cell biological analyses to assess effects of DEXA on GBM cells. We tested the efficacy of tyrosine kinase inhibitors in vitro and in vivo. (3) Results: We confirm in our patient cohort that administration of DEXA correlates with worse overall survival and shorter time to relapse. In GBM cells and glioma stem-like cells (GSCs) DEXA down-regulates genes controlling G2/M and mitotic-spindle checkpoints, and it enables cells to override the spindle assembly checkpoint (SAC). Concurrently, DEXA up-regulates Platelet Derived Growth Factor Receptor (PDGFR) signalling, which stimulates expression of anti-apoptotic regulators BCL2L1 and MCL1, required for survival during extended mitosis. Importantly, the protective potential of DEXA is dependent on intact tyrosine kinase signalling and ponatinib, sunitinib and dasatinib, all effectively overcome the radio-protective and pro-proliferative activity of DEXA. Moreover, we discovered that DEXA-induced signalling creates a therapeutic vulnerability for sunitinib in GSCs and GBM cells in vitro and in vivo. (4) Conclusions: Our results reveal a novel DEXA-induced mechanism in GBM cells and provide a rationale for revisiting the use of tyrosine kinase inhibitors for the treatment of GBM.

18.
Nat Commun ; 11(1): 2977, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32532995

ABSTRACT

Independent scientific achievements have led to the discovery of aberrant splicing patterns in oncogenesis, while more recent advances have uncovered novel gene fusions involving neurotrophic tyrosine receptor kinases (NTRKs) in gliomas. The exploration of NTRK splice variants in normal and neoplastic brain provides an intersection of these two rapidly evolving fields. Tropomyosin receptor kinase B (TrkB), encoded NTRK2, is known for critical roles in neuronal survival, differentiation, molecular properties associated with memory, and exhibits intricate splicing patterns and post-translational modifications. Here, we show a role for a truncated NTRK2 splice variant, TrkB.T1, in human glioma. TrkB.T1 enhances PDGF-driven gliomas in vivo, augments PDGF-induced Akt and STAT3 signaling in vitro, while next generation sequencing broadly implicates TrkB.T1 in the PI3K signaling cascades in a ligand-independent fashion. These TrkB.T1 findings highlight the importance of expanding upon whole gene and gene fusion analyses to include splice variants in basic and translational neuro-oncology research.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Membrane Glycoproteins/genetics , Oncogenes/genetics , RNA Isoforms/genetics , RNA Splicing , Receptor, trkB/genetics , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis/genetics , Cells, Cultured , Gene Expression Profiling , Gene Ontology , Glioma/metabolism , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Membrane Glycoproteins/metabolism , Mice , NIH 3T3 Cells , Neural Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RNA Isoforms/metabolism , Receptor, trkB/metabolism , Signal Transduction/genetics
19.
Nat Commun ; 11(1): 3883, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753598

ABSTRACT

Temozolomide (TMZ) is an oral alkylating agent used for the treatment of glioblastoma and is now becoming a chemotherapeutic option in patients diagnosed with high-risk low-grade gliomas. The O-6-methylguanine-DNA methyltransferase (MGMT) is responsible for the direct repair of the main TMZ-induced toxic DNA adduct, the O6-Methylguanine lesion. MGMT promoter hypermethylation is currently the only known biomarker for TMZ response in glioblastoma patients. Here we show that a subset of recurrent gliomas carries MGMT genomic rearrangements that lead to MGMT overexpression, independently from changes in its promoter methylation. By leveraging the CRISPR/Cas9 technology we generated some of these MGMT rearrangements in glioma cells and demonstrated that the MGMT genomic rearrangements contribute to TMZ resistance both in vitro and in vivo. Lastly, we showed that such fusions can be detected in tumor-derived exosomes and could potentially represent an early detection marker of tumor recurrence in a subset of patients treated with TMZ.


Subject(s)
Brain Neoplasms/drug therapy , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Gene Rearrangement , Glioma/drug therapy , Neoplasm Recurrence, Local/genetics , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Aged , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , DNA Adducts/drug effects , DNA Adducts/metabolism , DNA Methylation , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Male , Mice , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Promoter Regions, Genetic/genetics , RNA-Seq , Temozolomide/therapeutic use , Tumor Suppressor Proteins/metabolism , Up-Regulation , Whole Genome Sequencing , Xenograft Model Antitumor Assays , Young Adult
20.
Nat Commun ; 10(1): 2416, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186412

ABSTRACT

Cancer response to immunotherapy depends on the infiltration of CD8+ T cells and the presence of tumor-associated macrophages within tumors. Still, little is known about the determinants of these factors. We show that LIF assumes a crucial role in the regulation of CD8+ T cell tumor infiltration, while promoting the presence of protumoral tumor-associated macrophages. We observe that the blockade of LIF in tumors expressing high levels of LIF decreases CD206, CD163 and CCL2 and induces CXCL9 expression in tumor-associated macrophages. The blockade of LIF releases the epigenetic silencing of CXCL9 triggering CD8+ T cell tumor infiltration. The combination of LIF neutralizing antibodies with the inhibition of the PD1 immune checkpoint promotes tumor regression, immunological memory and an increase in overall survival.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL9/metabolism , Leukemia Inhibitory Factor/immunology , Macrophages/immunology , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Neutralizing/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL2/metabolism , Epigenesis, Genetic , Humans , Immunologic Memory , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, SCID , Neoplasm Transplantation , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL