Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 640
Filter
1.
Nat Commun ; 15(1): 6775, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117669

ABSTRACT

Structural variation heavily influences the molecular landscape of cancer, in part by impacting DNA methylation-mediated transcriptional regulation. Here, using multi-omic datasets involving >2400 pediatric brain and central nervous system tumors of diverse histologies from the Children's Brain Tumor Network, we report hundreds of genes and associated CpG islands (CGIs) for which the nearby presence of somatic structural variant (SV) breakpoints is recurrently associated with altered expression or DNA methylation, respectively, including tumor suppressor genes ATRX and CDKN2A. Altered DNA methylation near enhancers associates with nearby somatic SV breakpoints, including MYC and MYCN. A subset of genes with SV-CGI methylation associations also have expression associations with patient survival, including BCOR, TERT, RCOR2, and PDLIM4. DNA methylation changes in recurrent or progressive tumors compared to the initial tumor within the same patient can predict survival in pediatric and adult cancers. Our comprehensive and pan-histology genomic analyses reveal mechanisms of noncoding alterations impacting cancer genes.


Subject(s)
Brain Neoplasms , CpG Islands , DNA Methylation , Gene Expression Regulation, Neoplastic , Humans , DNA Methylation/genetics , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , CpG Islands/genetics , Child , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Epigenome , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Male , Telomerase/genetics , Female
3.
Hum Pathol ; 150: 51-57, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909708

ABSTRACT

Pancreatic neuroendocrine tumors (PanNETs) comprise a heterogeneous group of neoplasms in terms of biological behavior. This study aims to develop a practical algorithm based on emerging biomarkers, including chromatin-remodeling molecules DAXX/ATRX/H3K36me3, in conjunction with established prognostic factors, such as WHO grade and size. In immunohistochemical analyses, 18 of the 111 (16.2%) primary PanNETs showed DAXX or ATRX loss in a mutually exclusive manner. DAXX/ATRX loss was significantly correlated with higher recurrence risk and better predicted postoperative recurrence than WHO grade. We proposed a novel algorithm for stratifying patients with resectable PanNET into three groups according to recurrence risk: (A) WHO Grade 1 and ≤2 cm (very low-risk); for the others, (B) retained DAXX/ATRX (low-risk) and (C) DAXX/ATRX complete/heterogeneous loss (high-risk). Furthermore, we elucidated the intratumoral heterogeneities of PanNETs. Among cases with DAXX or ATRX loss, nine cases demonstrated heterogeneous loss of expression of DAXX/ATRX/H3K36me3. The majority of cases with DAXX/ATRX loss, either homogeneous or heterogeneous loss, showed uniform α-cell-like phenotype (ARX1+/PDX1-). In cases of metastatic or recurrent tumors, the expression pattern was identical to that observed in at least part of the primary tumor. In some instances, the expression pattern differed among different metastatic or recurrent tumors of the same patient. In summary, we propose a clinically useful and practical algorithm for postoperative recurrence risk stratification in PanNETs, by combining DAXX/ATRX status with WHO grade and size. Moreover, our findings highlighted the frequent spatiotemporal heterogeneity of chromatin-remodeling molecule expression in PanNETs with an α-cell phenotype, offering insights into tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Biomarkers, Tumor , Co-Repressor Proteins , Molecular Chaperones , Neoplasm Recurrence, Local , Neuroendocrine Tumors , Nuclear Proteins , Pancreatic Neoplasms , Phenotype , X-linked Nuclear Protein , Humans , X-linked Nuclear Protein/analysis , Pancreatic Neoplasms/pathology , Male , Female , Neoplasm Recurrence, Local/pathology , Neuroendocrine Tumors/pathology , Middle Aged , Aged , Adaptor Proteins, Signal Transducing/analysis , Biomarkers, Tumor/analysis , Nuclear Proteins/analysis , Adult , Risk Factors , Immunohistochemistry , Algorithms , Risk Assessment , Aged, 80 and over , Neoplasm Grading
5.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928515

ABSTRACT

Glioblastoma is the most aggressive tumor in the central nervous system, with a survival rate of less than 15 months despite multimodal therapy. Tumor recurrence frequently occurs after removal. Tumoral angiogenesis, the formation of neovessels, has a positive impact on tumor progression and invasion, although there are controversial results in the specialized literature regarding its impact on survival. This study aims to correlate the immunoexpression of angiogenesis markers (CD34, CD105) with the proliferation index Ki67 and p53 in primary and secondary glioblastomas. This retrospective study included 54 patients diagnosed with glioblastoma at the Pathology Department of County Emergency Clinical Hospital Târgu Mureș. Microvascular density was determined using CD34 and CD105 antibodies, and the results were correlated with the immunoexpression of p53, IDH1, ATRX and Ki67. The number of neoformed blood vessels varied among cases, characterized by different shapes and calibers, with endothelial cells showing modified morphology and moderate to marked pleomorphism. Neovessels with a glomeruloid aspect, associated with intense positivity for CD34 or CD105 in endothelial cells, were observed, characteristic of glioblastomas. Mean microvascular density values were higher for the CD34 marker in all cases, though there were no statistically significant differences compared to CD105. Mutant IDH1 and ATRX glioblastomas, wild-type p53 glioblastomas, and those with a Ki67 index above 20% showed a more abundant microvascular density, with statistical correlations not reaching significance. This study highlighted a variety of percentage intervals of microvascular density in primary and secondary glioblastomas using immunohistochemical markers CD34 and CD105, respectively, with no statistically significant correlation between evaluated microvascular density and p53 or Ki67.


Subject(s)
Brain Neoplasms , Glioblastoma , Isocitrate Dehydrogenase , Ki-67 Antigen , Microvascular Density , Neovascularization, Pathologic , Tumor Suppressor Protein p53 , X-linked Nuclear Protein , Humans , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/blood supply , Glioblastoma/genetics , Tumor Suppressor Protein p53/metabolism , Ki-67 Antigen/metabolism , Female , Middle Aged , Male , Aged , Adult , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , X-linked Nuclear Protein/metabolism , X-linked Nuclear Protein/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Retrospective Studies , Endoglin/metabolism , Endoglin/genetics , Antigens, CD34/metabolism , Biomarkers, Tumor/metabolism , Immunohistochemistry
6.
J Neurooncol ; 169(2): 287-297, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38874844

ABSTRACT

PURPOSE: To evaluate the performance of multi-pool Chemical exchange saturation transfer (CEST) MRI in prediction of glioma grade, isocitrate dehydrogenase (IDH) mutation, alpha-thalassemia/mental retardation syndrome X-linked (ATRX) loss and Ki-67 labeling index (LI), based on the fifth edition of the World Health Organization classification of central nervous system tumors (WHO CNS5). METHODS: 95 patients with adult-type diffuse gliomas were analyzed. The amide, direct water saturation (DS), nuclear Overhauser enhancement (NOE), semi-solid magnetization transfer (MT) and amine signals were derived using Lorentzian fitting, and asymmetry-based amide proton transfer-weighted (APTwasym) signal was calculated. The mean value of tumor region was measured and intergroup differences were estimated using student-t test. The receiver operating curve (ROC) and area under the curve (AUC) analysis were used to evaluate the diagnostic performance of signals and their combinations. Spearman correlation analysis was performed to evaluate tumor proliferation. RESULTS: The amide and DS signals were significantly higher in high-grade gliomas compared to low-grade gliomas, as well as in IDH-wildtype gliomas compared to IDH-mutant gliomas (all p < 0.001). The DS, MT and amine signals showed significantly differences between ATRX loss and retention in grade 2/3 IDH-mutant gliomas (all p < 0.05). The combination of signals showed the highest AUC in prediction of grade (0.857), IDH mutation (0.814) and ATRX loss (0.769). Additionally, the amide and DS signals were positively correlated with Ki-67 LI (both p < 0.001). CONCLUSION: Multi-pool CEST MRI demonstrated good potential to predict glioma grade, IDH mutation, ATRX loss and Ki-67 LI.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Magnetic Resonance Imaging , Mutation , Neoplasm Grading , Humans , Glioma/genetics , Glioma/diagnostic imaging , Glioma/metabolism , Glioma/pathology , Glioma/classification , Male , Female , Middle Aged , Adult , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Magnetic Resonance Imaging/methods , Isocitrate Dehydrogenase/genetics , Aged , Young Adult , Cell Proliferation , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Ki-67 Antigen/metabolism
7.
Nucleic Acids Res ; 52(11): 6472-6489, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38752489

ABSTRACT

Orphan nuclear receptors (NRs), such as COUP-TF1, COUP-TF2, EAR2, TR2 and TR4, are implicated in telomerase-negative cancers that maintain their telomeres through the alternative lengthening of telomeres (ALT) mechanism. However, how telomere association of orphan NRs is involved in ALT activation remains unclear. Here, we demonstrate that telomeric tethering of orphan NRs in human fibroblasts initiates formation of ALT-associated PML bodies (APBs) and features of ALT activity, including ALT telomere DNA synthesis, telomere sister chromatid exchange, and telomeric C-circle generation, suggesting de novo ALT induction. Overexpression of orphan NRs exacerbates ALT phenotypes in ALT cells, while their depletion limits ALT. Orphan NRs initiate ALT via the zinc finger protein 827, suggesting the involvement of chromatin structure alterations for ALT activation. Furthermore, we found that orphan NRs and deficiency of the ALT suppressor ATRX-DAXX complex operate in concert to promote ALT activation. Moreover, PML depletion by gene knockout or arsenic trioxide treatment inhibited ALT induction in fibroblasts and ALT cancer cells, suggesting that APB formation underlies the orphan NR-induced ALT activation. Importantly, arsenic trioxide administration abolished APB formation and features of ALT activity in ALT cancer cell line-derived mouse xenografts, suggesting its potential for further therapeutic development to treat ALT cancers.


Subject(s)
Fibroblasts , Promyelocytic Leukemia Protein , Telomere Homeostasis , Humans , Animals , Promyelocytic Leukemia Protein/metabolism , Promyelocytic Leukemia Protein/genetics , Mice , Fibroblasts/metabolism , Telomere/metabolism , Telomere/genetics , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Sister Chromatid Exchange , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Arsenic Trioxide/pharmacology , Molecular Chaperones
8.
Eur J Med Genet ; 69: 104949, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38797245

ABSTRACT

Variation in the non-coding genome is being increasingly recognized to be involved in monogenic disease etiology. However, the interpretation of non-coding variation is complicated by a lack of understanding of how non-coding genetic elements function. Additional lines of evidence are therefore needed to recognize non-coding variants as pathogenic. We here present a case where a collective body of evidence resulted in the identification and conclusive classification of a pathogenic deep intronic variant in ATRX. This report demonstrates the utility of a multi-platform approach in aiding the identification of pathogenic variants outside coding regions. Furthermore, it marks the first reported instance of a deep intronic pathogenic variant in ATRX.


Subject(s)
Introns , X-linked Nuclear Protein , Humans , X-linked Nuclear Protein/genetics , Male , Mutation , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Mental Retardation, X-Linked/diagnosis
9.
Sci Rep ; 14(1): 8510, 2024 04 12.
Article in English | MEDLINE | ID: mdl-38609433

ABSTRACT

Pancreatic neuroendocrine tumors (PanNETs) are a heterogeneous group of tumors that exhibit an unpredictable and broad spectrum of clinical presentations and biological aggressiveness. Surgical resection is still the only curative therapeutic option for localized PanNET, but the majority of patients are diagnosed at an advanced and metastatic stage with limited therapeutic options. Key factors limiting the development of new therapeutics are the extensive heterogeneity of PanNETs and the lack of appropriate clinically relevant models. In that context, genomic sequencing of human PanNETs revealed recurrent mutations and structural alterations in several tumor suppressors. Here, we demonstrated that combined loss of MEN1, ATRX, and PTEN, tumor suppressors commonly mutated in human PanNETs, triggers the development of high-grade pancreatic neuroendocrine tumors in mice. Histopathological evaluation and gene expression analyses of the developed tumors confirm the presence of PanNET hallmarks and significant overlap in gene expression patterns found in human disease. Thus, we postulate that the presented novel genetically defined mouse model is the first clinically relevant immunocompetent high-grade PanNET mouse model.


Subject(s)
Neuroendocrine Tumors , Pancreatic Neoplasms , Animals , Humans , Mice , Aggression , Chromosome Mapping , Disease Models, Animal , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , PTEN Phosphohydrolase/genetics , X-linked Nuclear Protein/genetics
10.
Sci Rep ; 14(1): 9427, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658591

ABSTRACT

Lower-grade gliomas (LGGs) exhibit highly variable clinical behaviors, while classic histology characteristics cannot accurately reflect the authentic biological behaviors, clinical outcomes, and prognosis of LGGs. In this study, we carried out analyses of whole exome sequencing, RNA sequencing and DNA methylation in primary vs. recurrent LGG samples, and also combined the multi-omics data to construct a prognostic prediction model. TCGA-LGG dataset was searched for LGG samples. 523 samples were used for whole exome sequencing analysis, 532 for transcriptional analysis, and 529 for DNA methylation analysis. LASSO regression was used to screen genes with significant association with LGG survival from the frequently mutated genes, differentially expressed genes, and differentially methylated genes, whereby a prediction model for prognosis of LGG was further constructed and validated. The most frequently mutated diver genes in LGGs were IDH1 (77%), TP53 (48%), ATRX (37%), etc. Top significantly up-regulated genes were C6orf15, DAO, MEOX2, etc., and top significantly down-regulated genes were DMBX1, GPR50, HMX2, etc. 2077 genes were more and 299 were less methylated in recurrent vs. primary LGG samples. Thirty-nine genes from the above analysis were included to establish a prediction model of survival, which showed that the high-score group had a very significantly shorter survival than the low-score group in both training and testing sets. ROC analysis showed that AUC was 0.817 for the training set and 0.819 for the testing set. This study will be beneficial to accurately predict the survival of LGGs to identify patients with poor prognosis to take specific treatment as early, which will help improve the treatment outcomes and prognosis of LGG.


Subject(s)
Brain Neoplasms , DNA Methylation , Glioma , Humans , Glioma/genetics , Glioma/pathology , Glioma/mortality , Prognosis , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Mutation , Male , Biomarkers, Tumor/genetics , Exome Sequencing , Neoplasm Grading , Gene Expression Profiling , X-linked Nuclear Protein/genetics , Middle Aged , Isocitrate Dehydrogenase/genetics , Multiomics
11.
Mol Cell ; 84(9): 1684-1698.e9, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38593805

ABSTRACT

The Bloom syndrome (BLM) helicase is critical for alternative lengthening of telomeres (ALT), a homology-directed repair (HDR)-mediated telomere maintenance mechanism that is prevalent in cancers of mesenchymal origin. The DNA substrates that BLM engages to direct telomere recombination during ALT remain unknown. Here, we determine that BLM helicase acts on lagging strand telomere intermediates that occur specifically in ALT-positive cells to assemble a replication-associated DNA damage response. Loss of ATRX was permissive for BLM localization to ALT telomeres in S and G2, commensurate with the appearance of telomere C-strand-specific single-stranded DNA (ssDNA). DNA2 nuclease deficiency increased 5'-flap formation in a BLM-dependent manner, while telomere C-strand, but not G-strand, nicks promoted ALT. These findings define the seminal events in the ALT DNA damage response, linking aberrant telomeric lagging strand DNA replication with a BLM-directed HDR mechanism that sustains telomere length in a subset of human cancers.


Subject(s)
DNA Damage , DNA Replication , RecQ Helicases , Telomere Homeostasis , Telomere , RecQ Helicases/metabolism , RecQ Helicases/genetics , Humans , Telomere/metabolism , Telomere/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Bloom Syndrome/enzymology , Bloom Syndrome/pathology , Cell Line, Tumor
12.
Nucleic Acids Res ; 52(9): 4950-4968, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38477352

ABSTRACT

Alterations in the tumor suppressor ATRX are recurrently observed in mesenchymal neoplasms. ATRX has multiple epigenetic functions including heterochromatin formation and maintenance and regulation of transcription through modulation of chromatin accessibility. Here, we show in murine mesenchymal progenitor cells (MPCs) that Atrx deficiency aberrantly activated mesenchymal differentiation programs. This includes adipogenic pathways where ATRX loss induced expression of adipogenic transcription factors and enhanced adipogenic differentiation in response to differentiation stimuli. These changes are linked to loss of heterochromatin near mesenchymal lineage genes together with increased chromatin accessibility and gains of active chromatin marks. We additionally observed depletion of H3K9me3 at transposable elements, which are derepressed including near mesenchymal genes where they could serve as regulatory elements. Finally, we demonstrated that loss of ATRX in a mesenchymal malignancy, undifferentiated pleomorphic sarcoma, results in similar epigenetic disruption and de-repression of transposable elements. Together, our results reveal a role for ATRX in maintaining epigenetic states and transcriptional repression in mesenchymal progenitors and tumor cells and in preventing aberrant differentiation in the progenitor context.


Subject(s)
Cell Differentiation , Heterochromatin , Mesenchymal Stem Cells , X-linked Nuclear Protein , Animals , Humans , Mice , Adipogenesis , DNA Transposable Elements/genetics , Epigenesis, Genetic , Heterochromatin/metabolism , Heterochromatin/genetics , Histones/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
13.
Neuro Oncol ; 26(8): 1479-1493, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38507506

ABSTRACT

BACKGROUND: H3 K27M-mutated gliomas were first described as a new grade 4 entity in the 2016 World Health Organization classification. Current studies have focused on its typical appearance in children and young adults, increasing the need to better understand the prognostic factors and impact of surgery on adults. Here, we report a multicentric study of this entity in adults. METHODS: We included molecularly confirmed H3 K27M-mutated glioma cases in patients ≥ 18 years diagnosed between 2016 and 2022. Clinical, radiological, and surgical features were analyzed. Univariate and multivariate analyses were performed to identify prognostic factors. RESULTS: Among 70 patients with a mean age of 36.1 years, the median overall survival (OS) was 13.6 ±â€…14 months. Gross-total resection was achieved in 14.3% of patients, whereas 30% had a subtotal resection and 54.3% a biopsy. Tumors located in telencephalon/diencephalon/myelencephalon were associated with a poorer OS, while a location in the mesencephalon/metencephalon showed a significantly longer OS (8.7 vs. 25.0 months, P = .007). Preoperative Karnofsky-Performance Score (KPS) ≤ 80 showed a reduced OS (4.2 vs. 18 months, P = .02). Furthermore, ATRX loss, found in 25.7%, was independently associated with an increased OS (31 vs. 8.3 months, P = .0029). Notably, patients undergoing resection showed no survival benefit over biopsy (12 vs. 11 months, P = .4006). CONCLUSIONS: The present study describes surgical features of H3 K27M-mutated glioma in adulthood in a large multicentric study. Our data reveal that ATRX status, location and KPS significantly impact OS in H3 K27M-mutated glioma. Importantly, our dataset indicates that resection does not offer a survival advantage over biopsy.


Subject(s)
Brain Neoplasms , Glioma , Histones , Mutation , X-linked Nuclear Protein , Humans , Male , Glioma/surgery , Glioma/genetics , Glioma/pathology , Glioma/mortality , Female , X-linked Nuclear Protein/genetics , Adult , Prognosis , Brain Neoplasms/surgery , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/mortality , Histones/genetics , Middle Aged , Young Adult , Survival Rate , Follow-Up Studies , Biomarkers, Tumor/genetics , Aged , Adolescent , Retrospective Studies
14.
J Imaging Inform Med ; 37(4): 1336-1345, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38378963

ABSTRACT

This study aimed to construct an imaging genomics nomogram based on intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) to predict the status of the alpha thalassemia/mental retardation syndrome X-linked (ATRX) gene in patients with brain gliomas. We retrospectively analyzed routine MR and IVIM-DWI data from 85 patients with pathologically confirmed brain gliomas from January 2017 to May 2023. The data were divided into a training set (N=61) and a test set (N=24) in a 7:3 ratio. Regions of interest (ROIs) of brain gliomas, including the solid tumor region (rCET), edema region (rE), and necrotic region (rNec), were delineated using 3D-Slicer software and projected onto the D, D*, and f sequences. A total of 1037 features were extracted from each ROI, resulting in 3111 features per patient. Age was incorporated in the calculation of the Radscore, and a clinical-imaging genomics combined model was constructed, from which a nomogram graph was generated. Separate models were built for the D, D*, and f parameters. The AUC value of the D parameter model was 0.97 (95% CI: 0.93-1.00) in the training set and 0.91 (95% CI: 0.79-1.00) in the validation set, which was significantly higher than that of the D* parameter model (0.90, 0.82) and the f parameter model (0.89, 0.91). The imaging genomics nomogram based on IVIM-DWI can effectively predict the ATRX gene status of patients with brain gliomas, with the D parameter showing the highest efficacy.


Subject(s)
Brain Neoplasms , Diffusion Magnetic Resonance Imaging , Glioma , Mutation , Nomograms , X-linked Nuclear Protein , Humans , Glioma/genetics , Glioma/diagnostic imaging , Glioma/pathology , Diffusion Magnetic Resonance Imaging/methods , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Male , Female , Adult , Middle Aged , Retrospective Studies , Young Adult , Adolescent , Aged
15.
Nat Commun ; 15(1): 730, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38272925

ABSTRACT

Stimulating the innate immune system has been explored as a therapeutic option for the treatment of gliomas. Inactivating mutations in ATRX, defining molecular alterations in IDH-mutant astrocytomas, have been implicated in dysfunctional immune signaling. However, little is known about the interplay between ATRX loss and IDH mutation on innate immunity. To explore this, we generated ATRX-deficient glioma models in the presence and absence of the IDH1R132H mutation. ATRX-deficient glioma cells are sensitive to dsRNA-based innate immune agonism and exhibit impaired lethality and increased T-cell infiltration in vivo. However, the presence of IDH1R132H dampens baseline expression of key innate immune genes and cytokines in a manner restored by genetic and pharmacological IDH1R132H inhibition. IDH1R132H co-expression does not interfere with the ATRX deficiency-mediated sensitivity to dsRNA. Thus, ATRX loss primes cells for recognition of dsRNA, while IDH1R132H reversibly masks this priming. This work reveals innate immunity as a therapeutic vulnerability of astrocytomas.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Humans , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , X-linked Nuclear Protein/genetics , Glioma/genetics , Glioma/metabolism , Astrocytoma/genetics , Mutation , Immunity, Innate/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism
16.
J Hum Genet ; 69(2): 101-105, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37904029

ABSTRACT

Partial duplications of genes can be challenging to detect and interpret and, therefore, likely represent an underreported cause of human disease. X-linked dominant variants in ATRX are associated with Alpha-thalassemia/impaired intellectual development syndrome, X-linked (ATR-X syndrome), a clinically heterogeneous disease generally presenting with intellectual disability, hypotonia, characteristic facies, genital anomalies, and alpha-thalassemia. We describe an affected male with a de novo hemizygous intragenic duplication of ~43.6 kb in ATRX, detected by research genome sequencing following non-diagnostic clinical testing. RNA sequencing and DNA methylation episignature analyses were central in variant interpretation, and this duplication was subsequently interpreted as disease-causing. This represents the smallest reported tandem duplication within ATRX associated with disease. This case demonstrates the diagnostic utility of integrating multiple omics technologies, which can ultimately lead to a definitive diagnosis for rare disease patients.


Subject(s)
Intellectual Disability , Mental Retardation, X-Linked , alpha-Thalassemia , Humans , Male , alpha-Thalassemia/diagnosis , alpha-Thalassemia/genetics , X-linked Nuclear Protein/genetics , DNA Copy Number Variations/genetics , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics
17.
J Clin Pathol ; 77(2): 82-86, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-37890990

ABSTRACT

Telomere maintenance and elongation allows cells to gain replicative immortality and evade cellular senescence during cancer development. While most cancers use telomerase to maintain telomere lengths, a subset of cancers engage the alternative lengthening of telomeres (ALT) pathway for telomere maintenance. ALT is present in 5%-10% of all cancers, although the prevalence is dramatically higher in certain cancer types, including complex karyotype sarcomas, isocitrate dehydrogenase-mutant astrocytoma (WHO grade II-IV), pancreatic neuroendocrine tumours, neuroblastoma and chromophobe hepatocellular carcinomas. ALT is maintained through a homology-directed DNA repair mechanism. Resembling break-induced replication, this aberrant process results in dramatic cell-to-cell telomere length heterogeneity, widespread chromosomal instability and chronic replication stress. Additionally, ALT-positive cancers frequently harbour inactivating mutations in either chromatin remodelling proteins (ATRX, DAXX and H3F3A) or DNA damage repair factors (SMARCAL1 and SLX4IP). ALT can readily be detected in tissue by assessing the presence of unique molecular characteristics, such as large ultrabright nuclear telomeric foci or partially single-stranded telomeric DNA circles (C-circles). Importantly, ALT has been validated as a robust diagnostic and prognostic biomarker for certain cancer types and may even be exploited as a therapeutic target via small molecular inhibitors and/or synthetic lethality approaches.


Subject(s)
Neoplasms , Telomerase , Humans , Telomere Homeostasis , X-linked Nuclear Protein/genetics , Telomerase/genetics , Telomere/genetics , Telomere/metabolism , Neoplasms/genetics , DNA Helicases/genetics , Carrier Proteins
18.
Nucleic Acids Res ; 52(3): 1136-1155, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38038252

ABSTRACT

Maintaining chromatin integrity at the repetitive non-coding DNA sequences underlying centromeres is crucial to prevent replicative stress, DNA breaks and genomic instability. The concerted action of transcriptional repressors, chromatin remodelling complexes and epigenetic factors controls transcription and chromatin structure in these regions. The histone chaperone complex ATRX/DAXX is involved in the establishment and maintenance of centromeric chromatin through the deposition of the histone variant H3.3. ATRX and DAXX have also evolved mutually-independent functions in transcription and chromatin dynamics. Here, using paediatric glioma and pancreatic neuroendocrine tumor cell lines, we identify a novel ATRX-independent function for DAXX in promoting genome stability by preventing transcription-associated R-loop accumulation and DNA double-strand break formation at centromeres. This function of DAXX required its interaction with histone H3.3 but was independent of H3.3 deposition and did not reflect a role in the repression of centromeric transcription. DAXX depletion mobilized BRCA1 at centromeres, in line with BRCA1 role in counteracting centromeric R-loop accumulation. Our results provide novel insights into the mechanisms protecting the human genome from chromosomal instability, as well as potential perspectives in the treatment of cancers with DAXX alterations.


Subject(s)
Centromere , DNA Breaks, Double-Stranded , Molecular Chaperones , Nuclear Proteins , R-Loop Structures , X-linked Nuclear Protein , Child , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Centromere/metabolism , Chromatin , Co-Repressor Proteins/metabolism , DNA , Histones/genetics , Histones/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
20.
Neuro Oncol ; 26(1): 55-67, 2024 01 05.
Article in English | MEDLINE | ID: mdl-37625115

ABSTRACT

BACKGROUND: Functional inactivation of ATRX characterizes large subgroups of malignant gliomas in adults and children. ATRX deficiency in glioma induces widespread chromatin remodeling, driving transcriptional shifts and oncogenic phenotypes. Effective strategies to therapeutically target these broad epigenomic sequelae remain undeveloped. METHODS: We utilized integrated multiomics and the Broad Institute Connectivity Map (CMAP) to identify drug candidates that could potentially revert ATRX-deficient transcriptional changes. We then employed disease-relevant experimental models to evaluate functional phenotypes, coupling these studies with epigenomic profiling to elucidate molecular mechanism(s). RESULTS: CMAP analysis and transcriptional/epigenomic profiling implicated the Class III HDAC Sirtuin2 (SIRT2) as a central mediator of ATRX-deficient cellular phenotypes and a driver of unfavorable prognosis in ATRX-deficient glioma. SIRT2 inhibitors reverted Atrx-deficient transcriptional signatures in murine neuroepithelial progenitor cells (mNPCs), impaired cell migration in Atrx/ATRX-deficient mNPCs and human glioma stem cells (GSCs), and increased expression of senescence markers in glioma models. Moreover, SIRT2 inhibition impaired growth and increased senescence in ATRX-deficient GSCs in vivo. These effects were accompanied by genome-wide shifts in enhancer-associated H3K27ac and H4K16ac marks, with the latter in particular demonstrating compelling transcriptional links to SIRT2-dependent phenotypic reversals. Motif analysis of these data identified the transcription factor KLF16 as a mediator of phenotype reversal in Atrx-deficient cells upon SIRT2 inhibition. CONCLUSIONS: Our findings indicate that SIRT2 inhibition selectively targets ATRX-deficient gliomas for senescence through global chromatin remodeling, while demonstrating more broadly a viable approach to combat complex epigenetic rewiring in cancer.


Subject(s)
Chromatin , Glioma , Adult , Child , Humans , Animals , Mice , Sirtuin 2/genetics , Sirtuin 2/metabolism , Glioma/pathology , X-linked Nuclear Protein/genetics , Kruppel-Like Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL