Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33758097

ABSTRACT

Most glioblastomas (GBMs) achieve cellular immortality by acquiring a mutation in the telomerase reverse transcriptase (TERT) promoter. TERT promoter mutations create a binding site for a GA binding protein (GABP) transcription factor complex, whose assembly at the promoter is associated with TERT reactivation and telomere maintenance. Here, we demonstrate increased binding of a specific GABPB1L-isoform-containing complex to the mutant TERT promoter. Furthermore, we find that TERT promoter mutant GBM cells, unlike wild-type cells, exhibit a critical near-term dependence on GABPB1L for proliferation, notably also posttumor establishment in vivo. Up-regulation of the protein paralogue GABPB2, which is normally expressed at very low levels, can rescue this dependence. More importantly, when combined with frontline temozolomide (TMZ) chemotherapy, inducible GABPB1L knockdown and the associated TERT reduction led to an impaired DNA damage response that resulted in profoundly reduced growth of intracranial GBM tumors. Together, these findings provide insights into the mechanism of cancer-specific TERT regulation, uncover rapid effects of GABPB1L-mediated TERT suppression in GBM maintenance, and establish GABPB1L inhibition in combination with chemotherapy as a therapeutic strategy for TERT promoter mutant GBM.


Subject(s)
Brain Neoplasms/genetics , GA-Binding Protein Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Telomerase/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Astrocytes , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA Damage/drug effects , Drug Resistance, Neoplasm/genetics , Female , GA-Binding Protein Transcription Factor/genetics , Gene Knockdown Techniques , Gene Knockout Techniques , Glioblastoma/drug therapy , Glioblastoma/pathology , HEK293 Cells , Humans , Mice , Mutation , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
2.
Cell Rep ; 40(12): 111344, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130485

ABSTRACT

Telomerase activation counteracts senescence and telomere erosion caused by uncontrolled proliferation. Epidermal growth factor receptor (EGFR) amplification drives proliferation while telomerase reverse transcriptase promoter (TERTp) mutations underlie telomerase reactivation through recruitment of GA-binding protein (GABP). EGFR amplification and TERTp mutations typically co-occur in glioblastoma, the most common and aggressive primary brain tumor. To determine if these two frequent alterations driving proliferation and immortality are functionally connected, we combine analyses of copy number, mRNA, and protein data from tumor tissue with pharmacologic and genetic perturbations. We demonstrate that proliferation arrest decreases TERT expression in a GABP-dependent manner and elucidate a critical proliferation-to-immortality pathway from EGFR to TERT expression selectively from the mutant TERTp through activation of AMP-mediated kinase (AMPK) and GABP upregulation. EGFR-AMPK signaling promotes telomerase activity and maintains telomere length. These results define how the tumor cell immortality mechanism keeps pace with persistent oncogene signaling and cell cycling.


Subject(s)
Glioblastoma , Telomerase , AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate , ErbB Receptors/genetics , ErbB Receptors/metabolism , GA-Binding Protein Transcription Factor/metabolism , Glioblastoma/genetics , Humans , Mutation/genetics , Oncogenes , RNA, Messenger , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism
3.
Neuro Oncol ; 24(12): 2063-2075, 2022 12 01.
Article in English | MEDLINE | ID: mdl-35325218

ABSTRACT

BACKGROUND: Heterozygous TERT (telomerase reverse transcriptase) promoter mutations (TPMs) facilitate TERT expression and are the most frequent mutation in glioblastoma (GBM). A recent analysis revealed this mutation is one of the earliest events in gliomagenesis. However, no appropriate human models have been engineered to study the role of this mutation in the initiation of these tumors. METHOD: We established GBM models by introducing the heterozygous TPM in human induced pluripotent stem cells (hiPSCs) using a two-step targeting approach in the context of GBM genetic alterations, CDKN2A/B and PTEN deletion, and EGFRvIII overexpression. The impact of the mutation was evaluated through the in vivo passage and in vitro experiment and analysis. RESULTS: Orthotopic injection of neuronal precursor cells (NPCs) derived from hiPSCs with the TPM into immunodeficient mice did not enhance tumorigenesis compared to TERT promoter wild type NPCs at initial in vivo passage presumably due to relatively long telomeres. However, the mutation recruited GA-Binding Protein and engendered low-level TERT expression resulting in enhanced tumorigenesis and maintenance of short telomeres upon secondary passage as observed in human GBM. These results provide the first insights regarding increased tumorigenesis upon introducing a TPM compared to isogenic controls without TPMs. CONCLUSION: Our novel GBM models presented the growth advantage of heterozygous TPMs for the first time in the context of GBM driver mutations relative to isogenic controls, thereby allowing for the identification and validation of TERT promoter-specific vulnerabilities in a genetically accurate background.


Subject(s)
Glioblastoma , Induced Pluripotent Stem Cells , Telomerase , Humans , Mice , Animals , Telomere Shortening/genetics , Telomerase/genetics , Telomere/genetics , Glioblastoma/genetics , Mutation , Carcinogenesis
4.
Neuro Oncol ; 23(4): 638-649, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33130899

ABSTRACT

BACKGROUND: Large-scale genome-wide association studies (GWAS) have implicated thousands of germline genetic variants in modulating individuals' risk to various diseases, including cancer. At least 25 risk loci have been identified for low-grade gliomas (LGGs), but their molecular functions remain largely unknown. METHODS: We hypothesized that GWAS loci contain causal single nucleotide polymorphisms (SNPs) that reside in accessible open chromatin regions and modulate the expression of target genes by perturbing the binding affinity of transcription factors (TFs). We performed an integrative analysis of genomic and epigenomic data from The Cancer Genome Atlas and other public repositories to identify candidate causal SNPs within linkage disequilibrium blocks of LGG GWAS loci. We assessed their potential regulatory role via in silico TF binding sequence perturbations, convolutional neural network trained on TF binding data, and simulated annealing-based interpretation methods. RESULTS: We built an interactive website (http://education.knoweng.org/alg3/) summarizing the functional footprinting of 280 variants in 25 LGG GWAS regions, providing rich information for further computational and experimental scrutiny. We identified as case studies PHLDB1 and SLC25A26 as candidate target genes of rs12803321 and rs11706832, respectively, and predicted the GWAS variant rs648044 to be the causal SNP modulating ZBTB16, a known tumor suppressor in multiple cancers. We showed that rs648044 likely perturbed the binding affinity of the TF MAFF, as supported by RNA interference and in vitro MAFF binding experiments. CONCLUSIONS: The identified candidate (causal SNP, target gene, TF) triplets and the accompanying resource will help accelerate our understanding of the molecular mechanisms underlying genetic risk factors for gliomas.


Subject(s)
Genome-Wide Association Study , Glioma , Amino Acid Transport Systems , Calcium-Binding Proteins , Genetic Predisposition to Disease , Glioma/genetics , Humans , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cancer Cell ; 34(3): 513-528.e8, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30205050

ABSTRACT

TERT promoter mutations reactivate telomerase, allowing for indefinite telomere maintenance and enabling cellular immortalization. These mutations specifically recruit the multimeric ETS factor GABP, which can form two functionally independent transcription factor species: a dimer or a tetramer. We show that genetic disruption of GABPß1L (ß1L), a tetramer-forming isoform of GABP that is dispensable for normal development, results in TERT silencing in a TERT promoter mutation-dependent manner. Reducing TERT expression by disrupting ß1L culminates in telomere loss and cell death exclusively in TERT promoter mutant cells. Orthotopic xenografting of ß1L-reduced, TERT promoter mutant glioblastoma cells rendered lower tumor burden and longer overall survival in mice. These results highlight the critical role of GABPß1L in enabling immortality in TERT promoter mutant glioblastoma.


Subject(s)
Brain Neoplasms/genetics , GA-Binding Protein Transcription Factor/metabolism , Glioblastoma/pathology , Promoter Regions, Genetic/genetics , Telomerase/genetics , Animals , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Female , GA-Binding Protein Transcription Factor/genetics , Gene Knockdown Techniques , Glioblastoma/genetics , Glioblastoma/mortality , Humans , Male , Mice , Mice, Nude , Mutation , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization/genetics , RNA, Small Interfering/metabolism , Survival Analysis , Telomerase/metabolism , Telomere/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL