Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Cells ; 13(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38727288

ABSTRACT

Glioblastoma (GBM) is a devastating brain cancer for which new effective therapies are urgently needed. GBM, after an initial response to current treatment regimens, develops therapeutic resistance, leading to rapid patient demise. Cancer cells exhibit an inherent elevation of endoplasmic reticulum (ER) stress due to uncontrolled growth and an unfavorable microenvironment, including hypoxia and nutrient deprivation. Cancer cells utilize the unfolded protein response (UPR) to maintain ER homeostasis, and failure of this response promotes cell death. In this study, as integrins are upregulated in cancer, we have evaluated the therapeutic potential of individually targeting all αß1 integrin subunits using RNA interference. We found that GBM cells are uniquely susceptible to silencing of integrin α3. Knockdown of α3-induced proapoptotic markers such as PARP cleavage and caspase 3 and 8 activation. Remarkably, we discovered a non-canonical function for α3 in mediating the maturation of integrin ß1. In its absence, generation of full length ß1 was reduced, immature ß1 accumulated, and the cells underwent elevated ER stress with upregulation of death receptor 5 (DR5) expression. Targeting α3 sensitized TRAIL-resistant GBM cancer cells to TRAIL-mediated apoptosis and led to growth inhibition. Our findings offer key new insights into integrin α3's role in GBM survival via the regulation of ER homeostasis and its value as a therapeutic target.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Glioblastoma , Integrin alpha3 , Integrin beta1 , TNF-Related Apoptosis-Inducing Ligand , Humans , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Apoptosis/genetics , Cell Line, Tumor , Integrin beta1/metabolism , Integrin beta1/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Integrin alpha3/metabolism , Integrin alpha3/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
2.
Cancer Res ; 84(5): 741-756, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38117484

ABSTRACT

Tumor adaptation or selection is thought to underlie therapy resistance in glioma. To investigate longitudinal epigenetic evolution of gliomas in response to therapeutic pressure, we performed an epigenomic analysis of 132 matched initial and recurrent tumors from patients with IDH-wildtype (IDHwt) and IDH-mutant (IDHmut) glioma. IDHwt gliomas showed a stable epigenome over time with relatively low levels of global methylation. The epigenome of IDHmut gliomas showed initial high levels of genome-wide DNA methylation that was progressively reduced to levels similar to those of IDHwt tumors. Integration of epigenomics, gene expression, and functional genomics identified HOXD13 as a master regulator of IDHmut astrocytoma evolution. Furthermore, relapse of IDHmut tumors was accompanied by histologic progression that was associated with survival, as validated in an independent cohort. Finally, the initial cell composition of the tumor microenvironment varied between IDHwt and IDHmut tumors and changed differentially following treatment, suggesting increased neoangiogenesis and T-cell infiltration upon treatment of IDHmut gliomas. This study provides one of the largest cohorts of paired longitudinal glioma samples with epigenomic, transcriptomic, and genomic profiling and suggests that treatment of IDHmut glioma is associated with epigenomic evolution toward an IDHwt-like phenotype. SIGNIFICANCE: Standard treatments are related to loss of DNA methylation in IDHmut glioma, resulting in epigenetic activation of genes associated with tumor progression and alterations in the microenvironment that resemble treatment-naïve IDHwt glioma.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Humans , Brain Neoplasms/pathology , Epigenesis, Genetic , Epigenomics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Neoplasm Recurrence, Local/genetics , Tumor Microenvironment
3.
Basic Clin Pharmacol Toxicol ; 133(4): 353-363, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37337931

ABSTRACT

Brain-specific angiogenesis inhibitor 3 (ADGRB3/BAI3) belongs to the family of adhesion G protein-coupled receptors. It is most highly expressed in the brain where it plays a role in synaptogenesis and synapse maintenance. Genome-wide association studies have implicated ADGRB3 in disorders such as schizophrenia and epilepsy. Somatic mutations in ADGRB3 have also been identified in cancer. To better understand the in vivo physiological role of ADGRB3, we used CRISPR/Cas9 editing to generate a mouse line with a 7-base pair deletion in Adgrb3 exon 10. Western blot analysis confirmed that homozygous mutants (Adgrb3∆7/∆7 ) lack full-length ADGRB3 expression. The mutant mice were viable and reproduced in Mendelian ratios but demonstrated reduced brain and body weights and deficits in social interaction. Measurements of locomotor function, olfaction, anxiety levels and prepulse inhibition were comparable between heterozygous and homozygous mutants and wild-type littermates. Since ADGRB3 is also expressed in organs such as lung and pancreas, this new mouse model will facilitate elucidation of ADGRB3's role in non-central nervous system-related functions. Finally, since somatic mutations in ADGRB3 were identified in patients with several cancer types, these mice can be used to determine whether loss of ADGRB3 function contributes to tumour development.


Subject(s)
Epilepsy , Neoplasms , Humans , Mice , Animals , Genome-Wide Association Study , Brain/metabolism , Receptors, G-Protein-Coupled/metabolism , Neoplasms/metabolism
4.
Cell ; 185(12): 2184-2199.e16, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35649412

ABSTRACT

The factors driving therapy resistance in diffuse glioma remain poorly understood. To identify treatment-associated cellular and genetic changes, we analyzed RNA and/or DNA sequencing data from the temporally separated tumor pairs of 304 adult patients with isocitrate dehydrogenase (IDH)-wild-type and IDH-mutant glioma. Tumors recurred in distinct manners that were dependent on IDH mutation status and attributable to changes in histological feature composition, somatic alterations, and microenvironment interactions. Hypermutation and acquired CDKN2A deletions were associated with an increase in proliferating neoplastic cells at recurrence in both glioma subtypes, reflecting active tumor growth. IDH-wild-type tumors were more invasive at recurrence, and their neoplastic cells exhibited increased expression of neuronal signaling programs that reflected a possible role for neuronal interactions in promoting glioma progression. Mesenchymal transition was associated with the presence of a myeloid cell state defined by specific ligand-receptor interactions with neoplastic cells. Collectively, these recurrence-associated phenotypes represent potential targets to alter disease progression.


Subject(s)
Brain Neoplasms , Glioma , Tumor Microenvironment , Adult , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Evolution, Molecular , Genes, p16 , Glioma/genetics , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Neoplasm Recurrence, Local
5.
Exp Neurol ; 351: 113994, 2022 05.
Article in English | MEDLINE | ID: mdl-35114205

ABSTRACT

The adhesion G protein-coupled receptor BAI1/ADGRB1 plays an important role in suppressing angiogenesis, mediating phagocytosis, and acting as a brain tumor suppressor. BAI1 is also a critical regulator of dendritic spine and excitatory synapse development and interacts with several autism-relevant proteins. However, little is known about the relationship between altered BAI1 function and clinically relevant phenotypes. Therefore, we studied the effect of reduced expression of full length Bai1 on behavior, seizure susceptibility, and brain morphology in Adgrb1 mutant mice. We compared homozygous (Adgrb1-/-), heterozygous (Adgrb1+/-), and wild-type (WT) littermates using a battery of tests to assess social behavior, anxiety, repetitive behavior, locomotor function, and seizure susceptibility. We found that Adgrb1-/- mice showed significant social behavior deficits and increased vulnerability to seizures. Adgrb1-/- mice also showed delayed growth and reduced brain weight. Furthermore, reduced neuron density and increased apoptosis during brain development were observed in the hippocampus of Adgrb1-/- mice, while levels of astrogliosis and microgliosis were comparable to WT littermates. These results show that reduced levels of full length Bai1 is associated with a broader range of clinically relevant phenotypes than previously reported.


Subject(s)
Angiogenic Proteins/metabolism , Receptors, G-Protein-Coupled , Angiogenic Proteins/genetics , Animals , Brain/metabolism , Hippocampus/metabolism , Mice , Receptors, G-Protein-Coupled/genetics , Seizures/genetics , Seizures/metabolism
6.
Neurooncol Adv ; 3(1): vdab088, 2021.
Article in English | MEDLINE | ID: mdl-34409295

ABSTRACT

BACKGROUND: Although IDH-mutant tumors aggregate to the frontotemporal regions, the clustering pattern of IDH-wildtype tumors is less clear. As voxel-based lesion-symptom mapping (VLSM) has several limitations for solid lesion mapping, a new technique, whole-lesion phenotype analysis (WLPA), is developed. We utilize WLPA to assess spatial clustering of tumors with IDH mutation from The Cancer Genome Atlas and The Cancer Imaging Archive. METHODS: The degree of tumor clustering segmented from T1 weighted images is measured to every other tumor by a function of lesion similarity to each other via the Hausdorff distance. Each tumor is ranked according to the degree to which its neighboring tumors show identical phenotypes, and through a permutation technique, significant tumors are determined. VLSM was applied through a previously described method. RESULTS: A total of 244 patients of mixed-grade gliomas (WHO II-IV) are analyzed, of which 150 were IDH-wildtype and 139 were glioblastomas. VLSM identifies frontal lobe regions that are more likely associated with the presence of IDH mutation but no regions where IDH-wildtype was more likely to be present. WLPA identifies both IDH-mutant and -wildtype tumors exhibit statistically significant spatial clustering. CONCLUSION: WLPA may provide additional statistical power when compared with VLSM without making several potentially erroneous assumptions. WLPA identifies tumors most likely to exhibit particular phenotypes, rather than producing anatomical maps, and may be used in conjunction with VLSM to understand the relationship between tumor morphology and biologically relevant tumor phenotypes.

7.
Acta Neuropathol ; 142(5): 859-871, 2021 11.
Article in English | MEDLINE | ID: mdl-34409497

ABSTRACT

Medulloblastoma, a common pediatric malignant central nervous system tumour, represent a small proportion of brain tumours in adults. Previously it has been shown that in adults, Sonic Hedgehog (SHH)-activated tumours predominate, with Wingless-type (WNT) and Group 4 being less common, but molecular risk stratification remains a challenge. We performed an integrated analysis consisting of genome-wide methylation profiling, copy number profiling, somatic nucleotide variants and correlation of clinical variables across a cohort of 191 adult medulloblastoma cases identified through the Medulloblastoma Advanced Genomics International Consortium. We identified 30 WNT, 112 SHH, 6 Group 3, and 41 Group 4 tumours. Patients with SHH tumours were significantly older at diagnosis compared to other subgroups (p < 0.0001). Five-year progression-free survival (PFS) for WNT, SHH, Group 3, and Group 4 tumours was 64.4 (48.0-86.5), 61.9% (51.6-74.2), 80.0% (95% CI 51.6-100.0), and 44.9% (95% CI 28.6-70.7), respectively (p = 0.06). None of the clinical variables (age, sex, metastatic status, extent of resection, chemotherapy, radiotherapy) were associated with subgroup-specific PFS. Survival among patients with SHH tumours was significantly worse for cases with chromosome 3p loss (HR 2.9, 95% CI 1.1-7.6; p = 0.02), chromosome 10q loss (HR 4.6, 95% CI 2.3-9.4; p < 0.0001), chromosome 17p loss (HR 2.3, 95% CI 1.1-4.8; p = 0.02), and PTCH1 mutations (HR 2.6, 95% CI 1.1-6.2; p = 0.04). The prognostic significance of 3p loss and 10q loss persisted in multivariable regression models. For Group 4 tumours, chromosome 8 loss was strongly associated with improved survival, which was validated in a non-overlapping cohort (combined cohort HR 0.2, 95% CI 0.1-0.7; p = 0.007). Unlike in pediatric medulloblastoma, whole chromosome 11 loss in Group 4 and chromosome 14q loss in SHH was not associated with improved survival, where MYCN, GLI2 and MYC amplification were rare. In sum, we report unique subgroup-specific cytogenetic features of adult medulloblastoma, which are distinct from those in younger patients, and correlate with survival disparities. Our findings suggest that clinical trials that incorporate new strategies tailored to high-risk adult medulloblastoma patients are urgently needed.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Adolescent , Adult , Biomarkers, Tumor/genetics , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Cohort Studies , Female , Humans , Male , Medulloblastoma/mortality , Medulloblastoma/pathology , Progression-Free Survival , Risk Factors , Young Adult
8.
Oncogene ; 40(33): 5182-5191, 2021 08.
Article in English | MEDLINE | ID: mdl-34218269

ABSTRACT

Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, and patients that develop metastases (~50%) survive <1 year, highlighting the urgent need for new therapies. TCGA has recently revealed that a hypoxia gene signature is associated with poor UM patient prognosis. Here we show that expression of hypoxia-regulated collagen prolyl-4-hydroxylase genes P4HA1 and P4HA2 is significantly upregulated in UM patients with metastatic disease and correlates with poor prognosis, suggesting these enzymes might be key tumor drivers. We targeted hypoxia-induced expression of P4HA1/2 in UM with KCN1, a hypoxia inducible factor-1 (HIF-1) pathway inhibitor and found potent inhibition of primary and metastatic disease and extension of animal survival, without overt side effects. At the molecular level, KCN1 antagonized hypoxia-induced expression of P4HA1 and P4HA2, which regulate collagen maturation and deposition in the extracellular matrix. The treatment decreased prolyl hydroxylation, induced proteolytic cleavage and rendered a disordered structure to collagen VI, the main collagen produced by UM, and reduced UM cell invasion. Together, these data demonstrate that extracellular collagen matrix formation can be targeted in UM by inhibiting hypoxia-induced P4HA1 and P4HA2 expression, warranting further development of this strategy in patients with uveal melanoma.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Extracellular Matrix , Humans , Hydroxylation , Melanoma , Transcriptional Activation , Up-Regulation , Uveal Neoplasms
9.
Genome Biol ; 22(1): 125, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33926529

ABSTRACT

BACKGROUND: Medulloblastoma (MB) is the most common malignant pediatric brain tumor that originates in the cerebellum and brainstem. Frequent somatic mutations and deregulated expression of epigenetic regulators in MB highlight the substantial role of epigenetic alterations. 5-hydroxymethylcytosine (5hmC) is a highly abundant cytosine modification in the developing cerebellum and is regulated by ten-eleven translocation (TET) enzymes. RESULTS: We investigate the alterations of 5hmC and TET enzymes in MB and their significance to cerebellar cancer formation. We show total abundance of 5hmC is reduced in MB, but identify significant enrichment of MB-specific 5hmC marks at regulatory regions of genes implicated in stem-like properties and Nanog-binding motifs. While TET1 and TET2 levels are high in MBs, only knockout of Tet1 in the smoothened (SmoA1) mouse model attenuates uncontrolled proliferation, leading to a favorable prognosis. The pharmacological Tet1 inhibition reduces cell viability and platelet-derived growth factor signaling pathway-associated genes. CONCLUSIONS: These results together suggest a potential key role of 5hmC and indicate an oncogenic nature for TET1 in MB tumorigenesis, suggesting it as a potential therapeutic target for MBs.


Subject(s)
Disease Susceptibility , Medulloblastoma/etiology , Medulloblastoma/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Biomarkers, Tumor , Computational Biology/methods , CpG Islands , DNA Methylation , Databases, Nucleic Acid , Disease Models, Animal , Disease Progression , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Medulloblastoma/mortality , Medulloblastoma/pathology , Mice , Mice, Transgenic , Nucleotide Motifs , Prognosis
10.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33720050

ABSTRACT

Glioblastoma (GBM) is composed of heterogeneous tumor cell populations, including those with stem cell properties, termed glioma stem cells (GSCs). GSCs are innately less radiation sensitive than the tumor bulk and are believed to drive GBM formation and recurrence after repeated irradiation. However, it is unclear how GSCs adapt to escape the toxicity of repeated irradiation used in clinical practice. To identify important mediators of adaptive radioresistance in GBM, we generated radioresistant human and mouse GSCs by exposing them to repeat cycles of irradiation. Surviving subpopulations acquired strong radioresistance in vivo, which was accompanied by a reduction in cell proliferation and an increase in cell-cell adhesion and N-cadherin expression. Increasing N-cadherin expression rendered parental GSCs radioresistant, reduced their proliferation, and increased their stemness and intercellular adhesive properties. Conversely, radioresistant GSCs lost their acquired phenotypes upon CRISPR/Cas9-mediated knockout of N-cadherin. Mechanistically, elevated N-cadherin expression resulted in the accumulation of ß-catenin at the cell surface, which suppressed Wnt/ß-catenin proliferative signaling, reduced neural differentiation, and protected against apoptosis through Clusterin secretion. N-cadherin upregulation was induced by radiation-induced IGF1 secretion, and the radiation resistance phenotype could be reverted with picropodophyllin, a clinically applicable blood-brain-barrier permeable IGF1 receptor inhibitor, supporting clinical translation.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Cadherins/metabolism , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Radiation Tolerance/physiology , Adaptation, Physiological , Animals , Antigens, CD/genetics , Apoptosis , Brain Neoplasms/pathology , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Clusterin/antagonists & inhibitors , Clusterin/genetics , Clusterin/metabolism , Female , Gene Knockout Techniques , Glioblastoma/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Radiation Tolerance/genetics , Up-Regulation , Wnt Signaling Pathway , Xenograft Model Antitumor Assays
11.
Nat Commun ; 12(1): 1749, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741928

ABSTRACT

Sonic hedgehog medulloblastoma encompasses a clinically and molecularly diverse group of cancers of the developing central nervous system. Here, we use unbiased sequencing of the transcriptome across a large cohort of 250 tumors to reveal differences among molecular subtypes of the disease, and demonstrate the previously unappreciated importance of non-coding RNA transcripts. We identify alterations within the cAMP dependent pathway (GNAS, PRKAR1A) which converge on GLI2 activity and show that 18% of tumors have a genetic event that directly targets the abundance and/or stability of MYCN. Furthermore, we discover an extensive network of fusions in focally amplified regions encompassing GLI2, and several loss-of-function fusions in tumor suppressor genes PTCH1, SUFU and NCOR1. Molecular convergence on a subset of genes by nucleotide variants, copy number aberrations, and gene fusions highlight the key roles of specific pathways in the pathogenesis of Sonic hedgehog medulloblastoma and open up opportunities for therapeutic intervention.


Subject(s)
Cerebellar Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Transcriptome , Adolescent , Adult , Child , Child, Preschool , Female , Gene Regulatory Networks , Genetic Variation , Humans , Infant , Male , Middle Aged , Signal Transduction/genetics , Young Adult
12.
Oncoscience ; 7(7-8): 47-48, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32923515
13.
Cell Rep Med ; 1(3)2020 06 23.
Article in English | MEDLINE | ID: mdl-32743560

ABSTRACT

Over the past decade, wingless-activated (WNT) medulloblastoma has been identified as a candidate for therapy de-escalation based on excellent survival; however, a paucity of relapses has precluded additional analyses of markers of relapse. To address this gap in knowledge, an international cohort of 93 molecularly confirmed WNT MB was assembled, where 5-year progression-free survival is 0.84 (95%, 0.763-0.925) with 15 relapsed individuals identified. Maintenance chemotherapy is identified as a strong predictor of relapse, with individuals receiving high doses of cyclophosphamide or ifosphamide having only one very late molecularly confirmed relapse (p = 0.032). The anatomical location of recurrence is metastatic in 12 of 15 relapses, with 8 of 12 metastatic relapses in the lateral ventricles. Maintenance chemotherapy, specifically cumulative cyclophosphamide doses, is a significant predictor of relapse across WNT MB. Future efforts to de-escalate therapy need to carefully consider not only the radiation dose but also the chemotherapy regimen and the propensity for metastatic relapses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adolescent , Biomarkers, Tumor/metabolism , Child , Cyclophosphamide/therapeutic use , Female , Humans , Ifosfamide/therapeutic use , Male , Medulloblastoma/metabolism , Middle Aged , Neoplasm Recurrence, Local/metabolism , Progression-Free Survival
14.
Clin Cancer Res ; 26(11): 2711-2724, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31969339

ABSTRACT

PURPOSE: Exploitation of altered glycosylation in cancer is a major goal for the design of new cancer therapy. Here, we designed a novel secreted chimeric signal peptide-Galectin-3 conjugate (sGal-3) and investigated its ability to induce cancer-specific cell death by targeting aberrantly N-glycosylated cell surface receptors on cancer cells. EXPERIMENTAL DESIGN: sGal-3 was genetically engineered from Gal-3 by extending its N-terminus with a noncleavable signal peptide from tissue plasminogen activator. sGal-3 killing ability was tested on normal and tumor cells in vitro and its antitumor activity was evaluated in subcutaneous lung cancer and orthotopic malignant glioma models. The mechanism of killing was investigated through assays detecting sGal-3 interaction with specific glycans on the surface of tumor cells and the elicited downstream proapoptotic signaling. RESULTS: We found sGal-3 preferentially binds to ß1 integrin on the surface of tumor cells due to aberrant N-glycosylation resulting from cancer-associated upregulation of several glycosyltransferases. This interaction induces potent cancer-specific death by triggering an oncoglycan-ß1/calpain/caspase-9 proapoptotic signaling cascade. sGal-3 could reduce the growth of subcutaneous lung cancers and malignant gliomas in brain, leading to increased animal survival. CONCLUSIONS: We demonstrate that sGal-3 kills aberrantly glycosylated tumor cells and antagonizes tumor growth through a novel integrin ß1-dependent cell-extrinsic apoptotic pathway. These findings provide proof-of-principle that aberrant N-oncoglycans represent valid cancer targets and support further translation of the chimeric sGal-3 peptide conjugate for cancer therapy.


Subject(s)
Apoptosis , Blood Proteins/metabolism , Galectins/metabolism , Glioma/drug therapy , Integrin beta1/metabolism , Peptide Fragments/pharmacology , Protein Sorting Signals , Animals , Blood Proteins/genetics , Cell Proliferation , Female , Galectins/genetics , Glioma/metabolism , Glioma/pathology , Glycosylation , Humans , Integrin beta1/genetics , Mice , Mice, Nude , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Oncogene ; 39(5): 1041-1048, 2020 01.
Article in English | MEDLINE | ID: mdl-31582835

ABSTRACT

Medulloblastoma (MB) is a malignant pediatric brain tumor for which new therapies are urgently needed. We demonstrate that treatment with EPZ-6438 (Tazemetostat), an enhancer of zeste homolog 2 (EZH2) inhibitor approved for clinical trials, blocks MB cell growth in vitro and in vivo, and prolongs survival in orthotopic xenograft models. We show that the therapeutic effect is dependent on epigenetic reactivation of adhesion G-protein-coupled receptor B1 (BAI1/ADGRB1), a tumor suppressor that controls p53 stability by blocking Mdm2. Histone 3 trimethylated on lysine 27 (H3K27me3), a marker of silent chromatin conformation is present at the ADGRB1 promoter, and inhibition of EZH2, the catalytic component of the Polycomb Repressive complex 2 (PRC2) that methylates H3K27, switches the gene into an active chromatin status and reactivates BAI1 expression. Mechanistically, targeting EZH2 promotes transition from H3K27me3 to H3K27ac at the promoter, recruits the C/EBPß (CREB-binding protein) and CBP transcription factors and activates ADGRB1 gene transcription. Taken together, our results identify key molecular players that regulate ADGRB1 gene expression in MB, demonstrate that reactivation of BAI1 expression underlies EPZ-6438 antitumorigenic action, and provide preclinical proof-of-principle evidence for targeting EZH2 in patients with MB.


Subject(s)
Angiogenic Proteins/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Medulloblastoma/pathology , Receptors, G-Protein-Coupled/genetics , Tumor Suppressor Protein p53/genetics , Angiogenic Proteins/deficiency , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Gene Silencing , Histones/metabolism , Humans , Methylation , Peptide Fragments/metabolism , Receptors, G-Protein-Coupled/deficiency , Sialoglycoproteins/metabolism
16.
Oncogene ; 39(5): 1165, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31676871

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

17.
Nature ; 574(7780): 707-711, 2019 10.
Article in English | MEDLINE | ID: mdl-31664194

ABSTRACT

In cancer, recurrent somatic single-nucleotide variants-which are rare in most paediatric cancers-are confined largely to protein-coding genes1-3. Here we report highly recurrent hotspot mutations (r.3A>G) of U1 spliceosomal small nuclear RNAs (snRNAs) in about 50% of Sonic hedgehog (SHH) medulloblastomas. These mutations were not present across other subgroups of medulloblastoma, and we identified these hotspot mutations in U1 snRNA in only <0.1% of 2,442 cancers, across 36 other tumour types. The mutations occur in 97% of adults (subtype SHHδ) and 25% of adolescents (subtype SHHα) with SHH medulloblastoma, but are largely absent from SHH medulloblastoma in infants. The U1 snRNA mutations occur in the 5' splice-site binding region, and snRNA-mutant tumours have significantly disrupted RNA splicing and an excess of 5' cryptic splicing events. Alternative splicing mediated by mutant U1 snRNA inactivates tumour-suppressor genes (PTCH1) and activates oncogenes (GLI2 and CCND2), and represents a target for therapy. These U1 snRNA mutations provide an example of highly recurrent and tissue-specific mutations of a non-protein-coding gene in cancer.


Subject(s)
Cerebellar Neoplasms/genetics , Hedgehog Proteins/genetics , Medulloblastoma/genetics , RNA, Small Nuclear/genetics , Adolescent , Adult , Alternative Splicing , Hedgehog Proteins/metabolism , Humans , Mutation , RNA Splice Sites , RNA Splicing
18.
Ann N Y Acad Sci ; 1456(1): 5-25, 2019 11.
Article in English | MEDLINE | ID: mdl-31168816

ABSTRACT

The adhesion class of G protein-coupled receptors (GPCRs) is the second largest family of GPCRs (33 members in humans). Adhesion GPCRs (aGPCRs) are defined by a large extracellular N-terminal region that is linked to a C-terminal seven transmembrane (7TM) domain via a GPCR-autoproteolysis inducing (GAIN) domain containing a GPCR proteolytic site (GPS). Most aGPCRs undergo autoproteolysis at the GPS motif, but the cleaved fragments stay closely associated, with the N-terminal fragment (NTF) bound to the 7TM of the C-terminal fragment (CTF). The NTFs of most aGPCRs contain domains known to be involved in cell-cell adhesion, while the CTFs are involved in classical G protein signaling, as well as other intracellular signaling. In this workshop report, we review the most recent findings on the biology, signaling mechanisms, and physiological functions of aGPCRs.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Humans , Receptors, G-Protein-Coupled/chemistry
20.
Sci Rep ; 9(1): 4437, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872606

ABSTRACT

CRISPR gene editing creates indels in targeted genes that are detected by genotyping. Separating PCR products generated from wild-type versus mutant alleles with small indels based on size is beyond the resolution capacity of regular agarose gel electrophoresis. To overcome this limitation, we developed a simple genotyping method that exploits the differential electrophoretic mobility of homoduplex versus heteroduplex DNA hybrids in high concentration agarose gels. First, the CRISPR target region is PCR amplified and homo- and hetero-duplexed amplicons formed during the last annealing cycle are separated by 4-6% agarose gel electrophoresis. WT/mutant heteroduplexes migrate more slowly and are distinguished from WT or mutant homoduplexes. Heterozygous alleles are immediately identified as they produce two distinct bands, while homozygous wild-type or mutant alleles yield a single band. To discriminate the latter, equal amounts of PCR products of homozygous samples are mixed with wild-type control samples, subjected to one denaturation/renaturation cycle and products are electrophoresed again. Samples from homozygous mutant alleles now produce two bands, while those from wild-type alleles yield single bands. This method is simple, fast and inexpensive and can identify indels >2 bp. in size in founder pups and genotype offspring in established transgenic mice colonies.


Subject(s)
CRISPR-Cas Systems , Electrophoresis, Agar Gel/methods , Gene Editing , INDEL Mutation , Membrane Proteins/genetics , NAV1.6 Voltage-Gated Sodium Channel/genetics , Nerve Tissue Proteins/genetics , Animals , Female , Genotype , Genotyping Techniques , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Transgenic , NAV1.6 Voltage-Gated Sodium Channel/chemistry , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...