Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncogene ; 40(16): 2830-2841, 2021 04.
Article in English | MEDLINE | ID: mdl-33731860

ABSTRACT

Adult pilocytic astrocytomas (PAs) have been regarded as indistinguishable from pediatric PAs in terms of genome-wide expression and methylation patterns. It has been unclear whether adult PAs arise early in life and remain asymptomatic until adulthood, or whether they develop during adulthood. We sought to determine the age and origin of adult human PAs using two types of "marks" in the genomic DNA. First, we analyzed the DNA methylation patterns of adult and pediatric PAs to distinguish between PAs of different anatomic locations (n = 257 PA and control brain tissues). Second, we measured the concentration of nuclear bomb test-derived 14C in genomic DNA (n = 14 cases), which indicates the time point of the formation of human cell populations. Our data suggest that adult and pediatric PAs developing in the infratentorial brain are closely related and potentially develop from precursor cells early in life, whereas supratentorial PAs might show age and location-specific differences.


Subject(s)
Astrocytoma/epidemiology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , Humans , Incidence , Infant , Infant, Newborn , Middle Aged , Young Adult
2.
Neuro Oncol ; 21(7): 867-877, 2019 07 11.
Article in English | MEDLINE | ID: mdl-30943283

ABSTRACT

BACKGROUND: Diffuse intrinsic pontine gliomas (DIPGs) are highly aggressive pediatric brain tumors that are characterized by a recurrent mutation (K27M) within the histone H3 encoding genes H3F3A and HIST1H3A/B/C. These mutations have been shown to induce a global reduction in the repressive histone modification H3K27me3, which together with widespread changes in DNA methylation patterns results in an extensive transcriptional reprogramming hampering the identification of single therapeutic targets based on a molecular rationale. METHODS: We applied a large-scale gene knockdown approach using a pooled short hairpin (sh)RNA library in combination with next-generation sequencing in order to identify DIPG-specific vulnerabilities. The therapeutic potential of specific inhibitors of candidate targets was validated in a secondary drug screen. RESULTS: We identified fibroblast growth factor receptor (FGFR) signaling and the serine/threonine protein phosphatase 2A (PP2A) as top depleted hits in patient-derived DIPG cell cultures and validated their lethal potential by FGF ligand depletion and genetic knockdown of the PP2A structural subunit PPP2R1A. Further, pharmacological inhibition of FGFR and PP2A signaling through ponatinib and LB-100 treatment, respectively, exhibited strong tumor-specific anti-proliferative and apoptotic activity in cultured DIPG cells. CONCLUSIONS: Our findings suggest FGFR and PP2A signaling as potential new therapeutic targets for the treatment of DIPGs.


Subject(s)
Biomarkers, Tumor/genetics , Brain Stem Neoplasms/genetics , Diffuse Intrinsic Pontine Glioma/genetics , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Apoptosis , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/pathology , Cell Proliferation , DNA Methylation , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/pathology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Library , High-Throughput Screening Assays , Humans , Protein Phosphatase 2/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Tumor Cells, Cultured
3.
Clin Cancer Res ; 25(6): 1851-1866, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30530705

ABSTRACT

PURPOSE: Pilocytic astrocytoma is the most common childhood brain tumor, characterized by constitutive MAPK activation. MAPK signaling induces oncogene-induced senescence (OIS), which may cause unpredictable growth behavior of pilocytic astrocytomas. The senescence-associated secretory phenotype (SASP) has been shown to regulate OIS, but its role in pilocytic astrocytoma remains unknown.Experimental Design: The patient-derived pilocytic astrocytoma cell culture model, DKFZ-BT66, was used to demonstrate presence of the SASP and analyze its impact on OIS in pilocytic astrocytoma. The model allows for doxycycline-inducible switching between proliferation and OIS. Both states were studied using gene expression profiling (GEP), Western blot, ELISA, and cell viability testing. Primary pilocytic astrocytoma tumors were analyzed by GEP and multiplex assay. RESULTS: SASP factors were upregulated in primary human and murine pilocytic astrocytoma and during OIS in DKFZ-BT66 cells. Conditioned medium induced growth arrest of proliferating pilocytic astrocytoma cells. The SASP factors IL1B and IL6 were upregulated in primary pilocytic astrocytoma, and both pathways were regulated during OIS in DKFZ-BT66. Stimulation with rIL1B but not rIL6 reduced growth of DKFZ-BT66 cells and induced the SASP. Anti-inflammatory treatment with dexamethasone induced regrowth of senescent cells and inhibited the SASP. Senescent DKFZ-BT66 cells responded to senolytic BCL2 inhibitors. High IL1B and SASP expression in pilocytic astrocytoma tumors was associated with favorable progression-free survival. CONCLUSIONS: We provide evidence for the SASP regulating OIS in pediatric pilocytic astrocytoma, with IL1B as a relevant mediator. SASP expression could enable prediction of progression in patients with pilocytic astrocytoma. Further investigation of the SASP driving the unpredictable growth of pilocytic astrocytomas, and its possible therapeutic application, is warranted.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Cellular Senescence , Interleukin-1beta/metabolism , Animals , Astrocytoma/mortality , Astrocytoma/surgery , Brain Neoplasms/mortality , Brain Neoplasms/surgery , Cell Proliferation , Child , Culture Media, Conditioned/metabolism , Datasets as Topic , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Male , Mice , Primary Cell Culture , Prognosis , Progression-Free Survival , Tumor Cells, Cultured
4.
Sci Rep ; 8(1): 17499, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504793

ABSTRACT

One key advantage of the CRISPR/Cas9 system in comparison with other gene editing approaches lies in its potential for multiplexing. Here, we describe an elaborate procedure that allows the assembly of multiple gRNA expression cassettes into a vector of choice within a single step, termed ASAP(Adaptable System for Assembly of multiplexed Plasmids)-cloning. We demonstrate the utility of ASAP-cloning for multiple CRISPR-mediated applications, including efficient multiplex gene editing, robust transcription activation and convenient analysis of Cas9 activity in the presence of multiple gRNAs.


Subject(s)
CRISPR-Cas Systems , Cloning, Molecular , Gene Editing , Genetic Vectors/genetics , Base Sequence , Cell Line, Tumor , Cloning, Molecular/methods , Gene Order , Genes, Reporter , Humans , RNA, Guide, Kinetoplastida/genetics , Sequence Analysis, DNA , Workflow
5.
EMBO Mol Med ; 10(8)2018 08.
Article in English | MEDLINE | ID: mdl-29973382

ABSTRACT

Most antidiabetic drugs treat disease symptoms rather than adipose tissue dysfunction as a key pathogenic cause in the metabolic syndrome and type 2 diabetes. Pharmacological targeting of adipose tissue through the nuclear receptor PPARg, as exemplified by glitazone treatments, mediates efficacious insulin sensitization. However, a better understanding of the context-specific PPARg responses is required for the development of novel approaches with reduced side effects. Here, we identified the transcriptional cofactor Cited4 as a target and mediator of rosiglitazone in human and murine adipocyte progenitor cells, where it promoted specific sets of the rosiglitazone-dependent transcriptional program. In mice, Cited4 was required for the proper induction of thermogenic expression by Rosi specifically in subcutaneous fat. This phenotype had high penetrance in females only and was not evident in beta-adrenergically stimulated browning. Intriguingly, this specific defect was associated with reduced capacity for systemic thermogenesis and compromised insulin sensitization upon therapeutic rosiglitazone treatment in female but not male mice. Our findings on Cited4 function reveal novel unexpected aspects of the pharmacological targeting of PPARg.


Subject(s)
Adipocytes/drug effects , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Rosiglitazone/therapeutic use , Transcription Factors/metabolism , Adipocytes/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Male , Mice , Molecular Targeted Therapy , PPAR gamma/metabolism , Sex Factors , Stem Cells/drug effects , Stem Cells/metabolism , Thermogenesis , Transcription Factors/biosynthesis , Transcription, Genetic/drug effects , Uncoupling Protein 1/biosynthesis
6.
Nat Commun ; 8: 14758, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28317875

ABSTRACT

Mutations in chromatin modifier genes are frequently associated with neurodevelopmental diseases. We herein demonstrate that the chromodomain helicase DNA-binding protein 7 (Chd7), frequently associated with CHARGE syndrome, is indispensable for normal cerebellar development. Genetic inactivation of Chd7 in cerebellar granule neuron progenitors leads to cerebellar hypoplasia in mice, due to the impairment of granule neuron differentiation, induction of apoptosis and abnormal localization of Purkinje cells, which closely recapitulates known clinical features in the cerebella of CHARGE patients. Combinatory molecular analyses reveal that Chd7 is required for the maintenance of open chromatin and thus activation of genes essential for granule neuron differentiation. We further demonstrate that both Chd7 and Top2b are necessary for the transcription of a set of long neuronal genes in cerebellar granule neurons. Altogether, our comprehensive analyses reveal a mechanism with chromatin remodellers governing brain development via controlling a core transcriptional programme for cell-specific differentiation.


Subject(s)
Brain/metabolism , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Neurons/metabolism , Animals , Brain/cytology , Brain/growth & development , Cerebellum/cytology , Cerebellum/growth & development , Cerebellum/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Humans , Mammals/genetics , Mammals/growth & development , Mammals/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/cytology
7.
Oncotarget ; 8(7): 11460-11479, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28002790

ABSTRACT

Pilocytic astrocytoma (PA) is the most frequent pediatric brain tumor. Activation of the MAPK pathway is well established as the oncogenic driver of the disease. It is most frequently caused by KIAA1549:BRAF fusions, and leads to oncogene induced senescence (OIS). OIS is thought to be a major reason for growth arrest of PA cells in vitro and in vivo, preventing establishment of PA cultures. Hence, valid preclinical models are currently very limited, but preclinical testing of new compounds is urgently needed. We transduced the PA short-term culture DKFZ-BT66 derived from the PA of a 2-year old patient with a doxycycline-inducible system coding for Simian Vacuolating Virus 40 Large T Antigen (SV40-TAg). SV40-TAg inhibits TP53/CDKN1A and CDKN2A/RB1, two pathways critical for OIS induction and maintenance. DNA methylation array and KIAA1549:BRAF fusion analysis confirmed pilocytic astrocytoma identity of DKFZ-BT66 cells after establishment. Readouts were analyzed in proliferating as well as senescent states, including cell counts, viability, cell cycle analysis, expression of SV40-Tag, CDKN2A (p16), CDKN1A (p21), and TP53 (p53) protein, and gene-expression profiling. Selected MAPK inhibitors (MAPKi) including clinically available MEK inhibitors (MEKi) were tested in vitro. Expression of SV40-TAg enabled the cells to bypass OIS and to resume proliferation with a mean doubling time of 45h allowing for propagation and long-term culture. Withdrawal of doxycycline led to an immediate decrease of SV40-TAg expression, appearance of senescent morphology, upregulation of CDKI proteins and a subsequent G1 growth arrest in line with the re-induction of senescence. DKFZ-BT66 cells still underwent replicative senescence that was overcome by TERT expression. Testing of a set of MAPKi revealed differential responses in DKFZ-BT66. MEKi efficiently inhibited MAPK signaling at clinically achievable concentrations, while BRAF V600E- and RAF Type II inhibitors showed paradoxical activation. Taken together, we have established the first patient-derived long term expandable PA cell line expressing the KIAA1549:BRAF-fusion suitable for preclinical drug testing.


Subject(s)
Astrocytoma , Brain Neoplasms , Cell Culture Techniques , Cell Line, Tumor , Cellular Senescence/physiology , Antigens, Polyomavirus Transforming/genetics , Blotting, Western , Cell Proliferation/physiology , Child, Preschool , Drug Screening Assays, Antitumor , Gene Expression Profiling , Humans , Male , Oncogene Proteins, Fusion/genetics , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Transcriptome , Transduction, Genetic
8.
Brief Funct Genomics ; 16(1): 25-33, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27273122

ABSTRACT

Advanced biological technologies allowing for genetic manipulation of the genome are increasingly being used to unravel the molecular pathogenesis of human diseases. The clustered regulatory interspaced short palindromic repeat/CRISPR-associated protein (CRISPR/Cas) technology started a revolution of this field owing to its flexibility and relative ease of use. Recently, application of the CRISPR/Cas9 system has been extended to in vivo approaches, leveraging its potential for human disease modeling. Particularly in oncological research, where genetic defects in somatic cells are tightly linked to etiology and pathological phenotypes, the CRISPR/Cas technology is being used to recapitulate various types of genetic aberrations. Here we review murine cancer models that have been developed via combining the CRISPR/Cas9 technology with in vivo somatic gene transfer approaches. Exploiting these methodological advances will further accelerate detailed investigations of tumor etiology and treatment.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Disease Models, Animal , Neoplasms/genetics , Animals , DNA Repair , Gene Deletion , Genetic Vectors , Mice , Neoplasms/pathology
9.
Nucleic Acids Res ; 45(1): e3, 2017 01 09.
Article in English | MEDLINE | ID: mdl-27614072

ABSTRACT

Successful RNAi applications depend on strategies allowing robust and persistent expression of minimal gene silencing triggers without perturbing endogenous gene expression. Here, we propose a novel avenue which is integration of a promoterless shmiRNA, i.e. a shRNA embedded in a micro-RNA (miRNA) scaffold, into an engineered genomic miRNA locus. For proof-of-concept, we used TALE or CRISPR/Cas9 nucleases to site-specifically integrate an anti-hepatitis C virus (HCV) shmiRNA into the liver-specific miR-122/hcr locus in hepatoma cells, with the aim to obtain cellular clones that are genetically protected against HCV infection. Using reporter assays, Northern blotting and qRT-PCR, we confirmed anti-HCV shmiRNA expression as well as miR-122 integrity and functionality in selected cellular progeny. Moreover, we employed a comprehensive battery of PCR, cDNA/miRNA profiling and whole genome sequencing analyses to validate targeted integration of a single shmiRNA molecule at the expected position, and to rule out deleterious effects on the genomes or transcriptomes of the engineered cells. Importantly, a subgenomic HCV replicon and a full-length reporter virus, but not a Dengue virus control, were significantly impaired in the modified cells. Our original combination of DNA engineering and RNAi expression technologies benefits numerous applications, from miRNA, genome and transgenesis research, to human gene therapy.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Genetic Engineering , Hepacivirus/genetics , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/genetics , Transcription Activator-Like Effector Nucleases/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Cell Line, Tumor , Disease Resistance/genetics , Endonucleases/genetics , Endonucleases/metabolism , Gene Editing , Gene Expression Profiling , Gene Expression Regulation , Genetic Loci , Genome, Human , HEK293 Cells , Hepatocytes/metabolism , Hepatocytes/virology , Host-Pathogen Interactions , Humans , MicroRNAs/metabolism , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , Transcription Activator-Like Effector Nucleases/metabolism , Virus Replication/genetics
11.
Nat Commun ; 6: 7391, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26067104

ABSTRACT

In vivo functional investigation of oncogenes using somatic gene transfer has been successfully exploited to validate their role in tumorigenesis. For tumour suppressor genes this has proven more challenging due to technical aspects. To provide a flexible and effective method for investigating somatic loss-of-function alterations and their influence on tumorigenesis, we have established CRISPR/Cas9-mediated somatic gene disruption, allowing for in vivo targeting of TSGs. Here we demonstrate the utility of this approach by deleting single (Ptch1) or multiple genes (Trp53, Pten, Nf1) in the mouse brain, resulting in the development of medulloblastoma and glioblastoma, respectively. Using whole-genome sequencing (WGS) we characterized the medulloblastoma-driving Ptch1 deletions in detail and show that no off-targets were detected in these tumours. This method provides a fast and convenient system for validating the emerging wealth of novel candidate tumour suppressor genes and the generation of faithful animal models of human cancer.


Subject(s)
Brain Neoplasms/genetics , CRISPR-Cas Systems , Disease Models, Animal , Gene Knockout Techniques/methods , Glioblastoma/genetics , Medulloblastoma/genetics , Animals , Brain Neoplasms/pathology , Gene Expression Profiling , Glioblastoma/pathology , Medulloblastoma/pathology , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neurofibromin 1/genetics , PTEN Phosphohydrolase/genetics , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics
12.
Cell Stem Cell ; 15(2): 185-98, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24835569

ABSTRACT

Cancer stem cells (CSCs) have been suggested as potential therapeutic targets for treating malignant tumors, but the in vivo supporting evidence is still missing. Using a GFP reporter driven by the promoter of the nuclear receptor tailless (Tlx), we demonstrate that Tlx(+) cells in primary brain tumors are mostly quiescent. Lineage tracing demonstrates that single Tlx(+) cells can self-renew and generate Tlx(-) tumor cells in primary tumors, suggesting that they are brain tumor stem cells (BTSCs). After introducing a BTSC-specific knock-out of the Tlx gene in primary mouse tumors, we observed a loss of self-renewal of BTSCs and prolongation of animal survival, accompanied by induction of essential signaling pathways mediating cell-cycle arrest, cell death, and neural differentiation. Our study demonstrates the feasibility of targeting glioblastomas and indicates the suitability of BTSCs as therapeutic targets, thereby supporting the CSC hypothesis.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Brain/pathology , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Survival , Glioma/metabolism , Green Fluorescent Proteins/metabolism , Humans , Mice , Neoplasm Transplantation , Nestin/metabolism , Neurons/cytology , Signal Transduction , Xenograft Model Antitumor Assays
13.
Cell ; 157(3): 525-7, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766799

ABSTRACT

Hierarchical cell state models, wherein a few stem-like tumor-propagating cells repopulate the tumor after therapy, are often invoked in cancer. Suvà et al. demonstrate a plastic developmental hierarchy in glioma cell populations by characterizing the epigenetic states of phenotypically distinct cells and identifying four factors sufficient to reprogram differentiated cells into a tumorigenic stem-like state.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Humans
14.
Cancer Cell ; 24(5): 660-72, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24183680

ABSTRACT

Two recurrent mutations, K27M and G34R/V, within histone variant H3.3 were recently identified in ∼50% of pHGGs. Both mutations define clinically and biologically distinct subgroups of pHGGs. Here, we provide further insight about the dominant-negative effect of K27M mutant H3.3, leading to a global reduction of the repressive histone mark H3K27me3. We demonstrate that this is caused by aberrant recruitment of the PRC2 complex to K27M mutant H3.3 and enzymatic inhibition of the H3K27me3-establishing methyltransferase EZH2. By performing chromatin immunoprecipitation followed by next-generation sequencing and whole-genome bisulfite sequencing in primary pHGGs, we show that reduced H3K27me3 levels and DNA hypomethylation act in concert to activate gene expression in K27M mutant pHGGs.


Subject(s)
Brain Neoplasms/genetics , Brain Stem Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Histones/genetics , Brain Neoplasms/metabolism , Brain Stem Neoplasms/metabolism , Cell Line, Tumor , Child , Epigenesis, Genetic , Genes, Dominant , Glioblastoma/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Methylation , Molecular Sequence Data , Mutation, Missense , Neoplasm Proteins , Polycomb Repressive Complex 2/metabolism , Protein Binding , Protein Processing, Post-Translational , Transcription Factors , Transcription, Genetic
15.
Nat Genet ; 45(8): 927-32, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23817572

ABSTRACT

Pilocytic astrocytoma, the most common childhood brain tumor, is typically associated with mitogen-activated protein kinase (MAPK) pathway alterations. Surgically inaccessible midline tumors are therapeutically challenging, showing sustained tendency for progression and often becoming a chronic disease with substantial morbidities. Here we describe whole-genome sequencing of 96 pilocytic astrocytomas, with matched RNA sequencing (n = 73), conducted by the International Cancer Genome Consortium (ICGC) PedBrain Tumor Project. We identified recurrent activating mutations in FGFR1 and PTPN11 and new NTRK2 fusion genes in non-cerebellar tumors. New BRAF-activating changes were also observed. MAPK pathway alterations affected all tumors analyzed, with no other significant mutations identified, indicating that pilocytic astrocytoma is predominantly a single-pathway disease. Notably, we identified the same FGFR1 mutations in a subset of H3F3A-mutated pediatric glioblastoma with additional alterations in the NF1 gene. Our findings thus identify new potential therapeutic targets in distinct subsets of pilocytic astrocytoma and childhood glioblastoma.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Mutation , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, trkB/genetics , Animals , Astrocytoma/metabolism , Base Sequence , Brain Neoplasms/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosome Breakpoints , Chromosomes, Human, Pair 6 , Chromosomes, Human, Pair 9 , Fibroblast Growth Factors/metabolism , Humans , MAP Kinase Signaling System , Mice , Models, Molecular , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Protein Conformation , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, trkB/metabolism
16.
Cell Mol Life Sci ; 69(11): 1799-811, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22159586

ABSTRACT

Pilocytic astrocytoma (PA) is the most common tumor of the pediatric central nervous system (CNS). A body of research over recent years has demonstrated a key role for mitogen-activated protein kinase (MAPK) pathway signaling in the development and behavior of PAs. Several mechanisms lead to activation of this pathway in PA, mostly in a mutually exclusive manner, with constitutive BRAF kinase activation subsequent to gene fusion being the most frequent. The high specificity of this fusion to PA when compared with other CNS tumors has diagnostic utility. In addition, the frequency of alteration of this key pathway provides an opportunity for molecularly targeted therapy in this tumor. Here, we review the current knowledge on mechanisms of MAPK activation in PA and some of the downstream consequences of this activation, which are now starting to be elucidated both in vitro and in vivo, as well as clinical considerations and possible future directions.


Subject(s)
Astrocytoma/metabolism , MAP Kinase Signaling System , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Disease Models, Animal , Gene Fusion , Humans , Mice , Models, Genetic , Neurofibromin 1/genetics , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
17.
J Clin Invest ; 121(4): 1344-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21403401

ABSTRACT

Pilocytic astrocytoma (PA) is the most common type of primary brain tumor in children and the second most frequent cancer in childhood. Children with incompletely resected PA represent a clinically challenging patient cohort for whom conventional adjuvant therapies are only moderately effective. This has produced high clinical demand for testing of new molecularly targeted treatments. However, the development of new therapeutics for PA has been hampered by the lack of an adequate in vivo tumor model. Recent studies have identified activation of MAPK signaling, mainly by oncogenic BRAF activation, as a hallmark genetic event in the pathogenesis of human PA. Using in vivo retroviral somatic gene transfer into mouse neural progenitor cells, we have shown here that ectopic expression of the activated BRAF kinase domain is sufficient to induce PA in mice. Further in vitro analyses demonstrated that overexpression of activated BRAF led to increased proliferation of primary mouse astrocytes that could be inhibited by treatment with the kinase inhibitor sorafenib. Our in vivo model for PA shows that the activated BRAF kinase domain is sufficient to induce PA and highlights its role as a potential therapeutic target.


Subject(s)
Astrocytoma/etiology , Brain Neoplasms/etiology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Animals , Animals, Newborn , Astrocytoma/enzymology , Astrocytoma/genetics , Astrocytoma/pathology , Base Sequence , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Primers/genetics , Disease Models, Animal , Enzyme Activation , Gene Transfer Techniques , Humans , MAP Kinase Signaling System , Mice , Mutation , Oncogenes , Protein Structure, Tertiary , Proto-Oncogene Proteins B-raf/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...