scholarly journals Genome-Wide DNA Methylation Analysis Reveals Epigenetic Dysregulation of MicroRNA-34A in TP53-Associated Cancer Susceptibility

2016 ◽  
Vol 34 (30) ◽  
pp. 3697-3704 ◽  
Author(s):  
Nardin Samuel ◽  
Gavin Wilson ◽  
Mathieu Lemire ◽  
Badr Id Said ◽  
Youliang Lou ◽  
...  

Purpose Although the link between mutant TP53 and human cancer is unequivocal, a significant knowledge gap exists in clinically actionable molecular targets in Li-Fraumeni syndrome (LFS), a highly penetrant cancer predisposition syndrome associated with germline mutations in TP53. This study surveyed the epigenome to identify functionally and clinically relevant novel genes implicated in LFS. Patients and Methods We performed genome-wide methylation analyses of peripheral blood leukocyte DNA in germline TP53 mutation carriers (n = 72) and individuals with TP53 wild type in whom histologically comparable malignancies developed (n = 111). Targeted bisulfite pyrosequencing was performed on a validation cohort of 30 TP53 mutation carriers and 46 patients with TP53 wild type, and candidate sites were evaluated in primary tumors from patients with LFS across multiple histologic tumor types. Results In 183 patients, distinct DNA methylation signatures were associated with deleterious TP53 mutations in peripheral blood leukocytes. TP53-associated DNA methylation marks occurred in genomic regions that harbored p53 binding sites and in genes encoding p53 pathway proteins. Moreover, loss-of-function TP53 mutations were significantly associated with differential methylation at the locus encoding microRNA miR-34A, a key component of the p53 regulatory network (adjusted P < .001), and validated in an independent patient cohort (n = 76, P < .001). Targeted bisulfite pyrosequencing demonstrated that miR-34A was inactivated by hypermethylation across many histologic types of primary tumors from patients with LFS. Moreover, miR-34A tumor hypermethylation was associated with decreased overall survival in a cohort of 29 patients with choroid plexus carcinomas, a characteristic LFS tumor (P < .05). Conclusion Epigenetic dysregulation of miR-34A may comprise an important path in TP53-associated cancer predisposition and represents a therapeutically actionable target with potential clinical relevance.

2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii32-iii32
Author(s):  
H Noor ◽  
R Rapkins ◽  
K McDonald

Abstract BACKGROUND Tumour Protein 53 (TP53) is a tumour suppressor gene that is mutated in at least 50% of human malignancies. The prevalence of TP53 mutation is much higher in astrocytomas with reports of up to 75% TP53 mutant cases. Rare cases of TP53 mutation also exist in oligodendroglial tumours (10–13%). P53 pathway is therefore an important factor in low-grade glioma tumorigenesis. Although the prognostic impact of TP53 mutations has been studied previously, no concrete concordance were reached between the studies. In this study, we investigated the prognostic effects of TP53 mutation in astrocytoma and oligodendroglioma. MATERIAL AND METHODS A cohort of 65 matched primary and recurrent fresh frozen tumours were sequenced to identify hotspot exons of TP53 mutation. Exons 1 to 10 were sequenced and pathogenic mutations were mostly predominant between Exons 4 and 8. The cohort was further expanded with 78 low grade glioma fresh frozen tissues and hotspot exons were sequenced. Selecting only the primary tumour from 65 matched tumours, a total of 50 Astrocytoma cases and 51 oligodendroglioma cases were analysed for prognostic effects of TP53. Only pathogenic TP53 mutations confirmed through COSMIC and NCBI databases were included in the over survival and progression-free survival analysis. RESULTS 62% (31/50) of astrocytomas and 16% (8/51) of oligodendrogliomas harboured pathogenic TP53 mutations. Pathogenic hotspot mutations in codon 273 (c.817 C>T and c.818 G>A) was prevalent in astrocytoma with 58% (18/31) of tumours with these mutations. TP53 mutation status was maintained between primary and recurrent tumours in 93% of cases. In astrocytoma, overall survival of TP53 mutant patients was longer compared to TP53 wild-type patients (p<0.01) but was not significant after adjusting for age, gender, grade and IDH1 mutation status. In contrast, astrocytoma patients with specific TP53 mutation in codon 273 showed significantly better survival compared to other TP53 mutant and TP53 wild-type patients combined (p<0.01) in our multivariate analysis. Time to first recurrence (progression-free survival) of TP53 mutant patients was significantly longer than TP53 wild-type patients (p<0.01) after adjustments were made, while TP53 mutation in codon 273 was not prognostic for progression-free survival. In oligodendroglioma patients, TP53 mutations did not significantly affect overall survival and progression-free survival. CONCLUSION In agreement with others, TP53 mutation is more prevalent in Astrocytoma and mutations in codon 273 are significantly associated with longer survival.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi5-vi5
Author(s):  
Wies Vallentgoed ◽  
Anneke Niers ◽  
Karin van Garderen ◽  
Martin van den Bent ◽  
Kaspar Draaisma ◽  
...  

Abstract The GLASS-NL consortium, was initiated to gain insight into the molecular mechanisms underlying glioma evolution and to identify markers of progression in IDH-mutant astrocytomas. Here, we present the first results of genome-wide DNA-methylation profiling of GLASS-NL samples. 110 adult patients were identified with an IDH-mutant astrocytoma at first diagnosis. All patients underwent a surgical resection of the tumor at least twice, separated by at least 6 months (median 40.9 months (IQR: 24.0, 64.7). In 37% and 18% of the cases, patients were treated with radiotherapy or chemotherapy respectively, before surgical resection of the recurrent tumor. DNA-methylation profiling was done on 235 samples from 103 patients (102 1st, 101 2nd, 29 3rd, and 3 4th resection). Copy number variations were also extracted from these data. Methylation classes were determined according to Capper et al. Overall survival (OS) was measured from date of first surgery. Of all primary tumors, the methylation-classifier assigned 85 (87%) to the low grade subclass and 10 (10%) to the high grade subclass. The relative proportion of high grade tumors increased ~three-fold at tumor recurrence (32/101, 32%) and even further in the second recurrence (15/29, 52%). Methylation classes were prognostic, both in primary and recurrent tumors. The overall DNA-methylation levels of recurrent samples was lower than that of primary samples. This difference is explained by the increased number of high grade samples at recurrence, since near identical DNA-methylation levels were observed in samples that remained low grade. In an unsupervised analysis, DNA-methylation data derived from primary and first recurrence samples of individual patients mostly (79%) cluster together. Recurrent samples that do not cluster with their primary tumor, form a separate group with relatively low genome-wide DNA-methylation. Our data demonstrate that methylation profiling identifies a shift towards a higher grade at tumor progression coinciding with reduced genome-wide DNA-methylation levels.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3283-3283
Author(s):  
Barbara Kantorova ◽  
Jitka Malcikova ◽  
Veronika Navrkalova ◽  
Jana Smardova ◽  
Kamila Brazdilova ◽  
...  

Abstract Introduction A presence of activating mutations in NOTCH1 gene has been recently associated with reduced survival and chemo-immunotherapy resistance in chronic lymphocytic leukemia (CLL). However, a prognostic significance of the NOTCH1 mutations with respect to TP53mutation status has not been fully explained yet. Methods An examined cohort included 409 patients with CLL enriched for high risk cases; in 121 patients consecutive samples were investigated. To determine the TP53 mutation status, a functional analysis of separated alleles in yeast (FASAY, exons 4-10) combined with direct sequencing was performed; the ambiguous cases were retested using an ultra-deep next generation sequencing (MiSeq platform; Illumina). The presence of NOTCH1 hotspot mutation (c.7544_7545delCT) was analyzed using direct sequencing complemented by allele-specific PCR in the selected samples. In several patients harboring concurrent NOTCH1 and TP53 mutations, single separated cancer cells were examined using multiplex PCR followed by direct sequencing. A correlation between mutation presence and patient overall survival, time to first treatment and other molecular and cytogenetic prognostic markers was assessed using Log-rank (Mantel-cox) test and Fisher's exact test, respectively. Results The NOTCH1 and TP53 mutations were detected in 16% (65/409) and 27% (110/409) of the examined patients, respectively; a coexistence of these mutations in the same blood samples was observed in 11% (19/175) of the mutated patients. The detected increased mutation frequency attributes to more unfavorable profile of the analyzed cohort; in the TP53-mutated patients missense substitutions predominated (75% of TP53 mutations). As expected, a significantly reduced overall survival in comparison to the wild-type cases (147 months) was observed in the NOTCH1-mutated (115 months; P = 0.0018), TP53-mutated (79 months; P < 0.0001) and NOTCH1-TP53-mutated patients (101 months; P = 0.0282). Since both NOTCH1 and TP53 mutations were strongly associated with an unmutated IGHV gene status (P < 0.0001 and P = 0.0007), we reanalyzed the IGHV-unmutated patients only and interestingly, the impact of simultaneous NOTCH1 and TP53 mutation presence on patient survival was missed in this case (P = 0.1478). On the other hand, in the NOTCH1 and/or TP53-mutated patients significantly reduced time to first treatment was identified as compared to the wild-type cases (41 months vs. 25 months in NOTCH1-mutated, P = 0.0075; 17 months in TP53-mutated, P < 0.0001; and 18 months in NOTCH1-TP53-mutated patients, P = 0.0003). The similar results were observed also in the subgroup of the IGHV-unmutated patients, with the exception of patients carrying sole NOTCH1 mutation (P = 0.2969). Moreover, in the NOTCH1-TP53-mutated patients an increased frequency of del(17p)(13.1) was found in comparison to the TP53-mutated patients only (72% vs. 56%); this cytogenetic defect was not detected in the patients with sole NOTCH1 mutation. Our results might indicate, that NOTCH1 mutation could preferentially co-selected with particular, less prognostic negative type of TP53 defects. Notably, in our cohort the NOTCH1 mutation predominated in the patients harboring truncating TP53 mutations localized in a C-terminal part of the TP53 gene behind the DNA-binding domain (P = 0.0128). Moreover, in one of the NOTCH1-TP53-mutated patients the analysis of separated cancer cells revealed a simultaneous presence of NOTCH1 mutation and TP53 in-frame deletion in the same CLL cell. In contrast, in the other examined NOTCH1-TP53-mutated patient the concurrent NOTCH1 mutation and TP53 missense substitution (with presumed negative impact on patient prognosis) were found in different CLL cells. Conclusions The parallel presence of NOTCH1 hotspot mutation might be detected in a significant proportion of TP53-mutated patients and it seems to be associated with less prognostic unfavorable TP53 mutations. Nevertheless, these preliminary data should be further confirmed in a large cohort of patients. This study was supported by projects VaVPI MSMT CR CZ.1.05/1.1.00/02.0068 of CEITEC, IGA MZ CR NT13493-4/2012, NT13519-4/2012 and CZ.1.07/2.3.00/30.0009. Disclosures Brychtova: Roche: Travel grants Other. Doubek:Roche: Travel grants Other.


Epigenomics ◽  
2021 ◽  
Author(s):  
Sonal Saxena ◽  
Sumana Choudhury ◽  
Pranay Amruth Maroju ◽  
Anuhya Anne ◽  
Lov Kumar ◽  
...  

Aim: To study the effects of DNMT1 overexpression on transcript levels of genes dysregulated in schizophrenia and on genome-wide methylation patterns. Materials & methods: Transcriptome and DNA methylome comparisons were made between R1 (wild-type) and Dnmt1tet/tet mouse embryonic stem cells and neurons overexpressing DNMT1. Genes dysregulated in both Dnmt1tet/tet cells and schizophrenia patients were studied further. Results & conclusions: About 50% of dysregulated genes in patients also showed altered transcript levels in Tet/Tet neurons in a DNA methylation-independent manner. These neurons unexpectedly showed genome-wide hypomethylation, increased transcript levels of Tet1 and Apobec 1-3 genes and increased activity and copy number of LINE-1 elements. The observed similarities between Tet/Tet neurons and schizophrenia brain samples reinforce DNMT1 overexpression as a risk factor.


2019 ◽  
Vol 20 (23) ◽  
pp. 5828 ◽  
Author(s):  
Stephanie Dias ◽  
Sumaiya Adam ◽  
Paul Rheeder ◽  
Johan Louw ◽  
Carmen Pheiffer

Increasing evidence implicate altered DNA methylation in the pathophysiology of gestational diabetes mellitus (GDM). This exploratory study probed the association between GDM and peripheral blood DNA methylation patterns in South African women. Genome-wide DNA methylation profiling was conducted in women with (n = 12) or without (n = 12) GDM using the Illumina Infinium HumanMethylationEPIC BeadChip array. Functional analysis of differentially methylated genes was conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. A total of 1046 CpG sites (associated with 939 genes) were differentially methylated between GDM and non-GDM groups. Enriched pathways included GDM-related pathways such as insulin resistance, glucose metabolism and inflammation. DNA methylation of the top five CpG loci showed distinct methylation patterns in GDM and non-GDM groups and was correlated with glucose concentrations. Of these, one CpG site mapped to the calmodulin-binding transcription activator 1 (CAMTA1) gene, which have been shown to regulate insulin production and secretion and may offer potential as an epigenetic biomarker in our population. Further validation using pyrosequencing and conducting longitudinal studies in large sample sizes and in different populations are required to investigate their candidacy as biomarkers of GDM.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 792-792
Author(s):  
Austin G Kulasekararaj ◽  
Alexander E Smith ◽  
Syed A Mian ◽  
Pramila Krishnamurthy ◽  
Azim M Mohamedali ◽  
...  

Abstract Abstract 792 Myelodysplastic syndromes are clinically and molecularly heterogeneous group of disorders with variable prognosis and propensity to leukaemic transformation. We analysed the incidence and impact of TP53 gene mutations in a large cohort of MDS patients (n=318) using next generation sequencing (454 FLX). The median age was 65 years (range 17–72 years) with a male to female ratio of 1.7:1.The median follow-up of this cohort was 18.7 months (range 1–74 months). Forty patients (12%) underwent disease modifying treatments (bone marrow transplant, intensive chemotherapy, 5-azacitidine and Lenalidomide) at the time of sample collection and hence survival analysis was censored at the date of such treatment. Twenty one (7%) has RA, MDS with isolated del 5q–26 (8%), RARS-13 (4%), RCMD/RCMD-RS-105 (32%), RAEB 1/2 −75 (24%), AML secondary to MDS-29 (9%), therapy related MDS (tMDS)-22(7%) and MDS/MPN 27 (9%). IPSS cytogenetic risk groups were; good risk-199(63%), intermediate −35 (11%) and poor risk-67 (21%) and cytogenetics failed in 17 patients (5%). The IPSS categories were, low risk: 71(24%), intermediate-1:101(32%), intermediate-2:58 (18%), high risk: 29 (9%) and 38 patients were not evaluable (proliferative CMML and MPD/MDS). Progression to AML occurred in 68 patients (20%). TP53mutations were observed in 30 (9.4%) of MDS patients. Twenty of 30 (67%) mutations were detected in int-2/high risk IPSS groups and were associated with complex cytogenetics (73%). Rest of the mutations were detected in low/int-1 IPSS patients with isolated 5q- abnormality [5 of 26, (19%)].Of the 22 cases with t MDS, 6 had TP53 mutations and 17/91 (19%) patients with RAEB and AML with trilineage dysplasia harboured TP53 mutations. Only one patient each with RCMD and MDS/MPD-U had TP53 mutation. Of the 39 mutations, 9 patients had two mutant P53 clones whereas 21 patients had single mutation. 30 of 36 (83%) mutations were located in the DNA-binding domain. Patients with therapy related MDS had increased likelihood (p<0.001) of having double mutations (5/22), while none of the patients with isolated 5q had double mutations (0/26).All except one patient (8 out of 9) with double mutations had a blast count of <10%.(p<0.001) The median clone size of TP53 mutations were 42 %( range 2.5–93%) and the median clone size of patients with double mutations was 82%.Nine patients with TP53 mutation had sequential samples tested at variable time intervals post 5-azacitidine treatment to analyze clone size and correlate with clinical responses. Four patients showed a marked decrease in clone size (24 to 3%,25:44% to 4:6%,35; 40% to 8; 17%78% to 15%,) associated with cytogenetic remission, one patient showed a increase in clone size (60% to 93%) at the time of leukaemic evolution and the remaining four patients had stable TP53 clones after 5-azacitidine. TP53 mutations strongly correlated with worse OS and PFS. The median OS for patients with wild type TP53 was not reached (NR,>66 months) compared with 9.7 months for patients with TP53 mutations (p<0.0001) and median PFS for patients with wild type TP53 was not reached (NR,>66 months) compared with 8.5 months for patients with TP53 mutations (p<0.0001). A cytogenetic sub-group analysis was performed in patients with complex cytogenetics with or without mutations of TP53, and showed patients with mutation had a worse OS (9.6 vs. 14.1 months, p<0.0001) and PFS (8.3 vs. 13.9 months, p<0.001) compared with those with wild type TP53. MDS with isolated deletion 5q harbouring a TP53 mutation(med OS 23 months) had significantly worse outcome compared with del5q patients without mutation(med OS 66 months)(p<0.002). In a multivariate analysis using co-variables: age (>55yr vs <55 yr), sex, WHO subtypes, IPSS risk category, transfusion dependency, presence or absence of mutations, TP53 was found to be the strongest predictor for OS (HR-3.78, p<0.0001, CI 2.37–6.20) and PFS (HR-3.22, p<0.0001, CI 1.97–5.26). This large single institution study confirms that TP53 mutations are an independent prognosticator of outcome in MDS. The adverse impact of TP53 persists after adjustment for cytogenetic risk and is of practical importance in evaluating the prognosis in patients with MDS. The relatively common occurrence of these mutations in different spectrums of MDS, ie isolated 5q- and complex cytogenetics, possibly implies two different mechanistic roles for p53 protein in disease evolution. Disclosures: Elebute: Alexion: Honoraria. Mufti:Celgene Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 870-870
Author(s):  
Nadja Blagitko-Dorfs ◽  
Pascal Schlosser ◽  
Rainer Claus ◽  
Tobias Ma ◽  
Katharina Götze ◽  
...  

Abstract Introduction: Treatment of acute myeloid leukemia (AML) in elderly patients remains challenging. Low-dose DNA hypomethylating agents are a therapeutic option in myelodysplastic syndromes and AML. However, the mechanism of action of hypomethylating agents and the role of induction of DNA hypomethylation in the clinical response is still unclear. To unravel the in vivoeffects of sequential cycles of decitabine, we set out to characterize methylomes of leukemic blasts, T cells (presumably not part of the malignant clone) and granulocytes before and during treatment of AML patients enrolled in the randomized phase II DECIDER clinical trial (NCT00867672). We developed a statistical model for longitudinal data analysis to identify the strongest hypomethylation response. Methods: Peripheral blood mononuclear cells (PBMC) from AML patients were collected before and during therapy (i.v. 20 mg/m2 decitabine for 5 days, with or without subsequent oral drug add-on). Leukemic blasts and T-cells were isolated using automatic magnetic sorting of cells (autoMACS) labelled with anti-human CD34, CD117 and CD3 MACS microbeads (Miltenyi Biotec), respectively. Granulocytes were isolated using dextran sedimentation. Cell type specific genome-wide DNA methylation profiles were obtained using Infinium Human Methylation 450 BeadChip arrays. Data were analyzed using R packages RnBeads applying beta mixture quantile dilation for normalization (Teschendorff et al. Bioinformatics, 29:189–196, 2013) and a modified version of NHMMfdr for multiple testing. Results: Peripheral blood blasts (median purity: 92%) were isolated from 20 patients, and T cells (median purity: 94%) from 26 patients before treatment and on days 4 and/or 8 and 15 of treatment cycle 1. From 10 patients, blasts and T cells were also collected during and/or after cycle 2. In total, until now 127 methylomes (46 blasts, 47 T cells, 34 granulocytes) were generated and used for mathematical modelling. Since the trial is still recruiting, genome-wide methylation was interpreted blinded to all clinical data including drug add-on (ATRA, valproic acid). First, the methylation dynamics of each individual CpG site described by a specified summary statistics were identified. Then, inter-probe distance and CpG annotation were incorporated to explain the dependence structure between CpG sites. In order to control the false discovery rate (FDR), we adapted a method proposed for differential DNA methylation (Kuan & Chiang, Biometrics 68: 774–783, 2012). The summary statistics for each CpG site were modelled to follow a non–homogeneous hidden Markov model. Statistical testing was validated by simulations revealing a very high discriminative power for affected CpGs even with very low methylation dynamics. Applying the model to blasts and T cells, extensive differences in the in vivomethylation changes became apparent. In blasts, 13% of CpG (59,920 CpGs of total 460,343 CpGs) showed significant DNA hypomethylation (Δβ>0.1, FDR<0.05) shared between patients by day 8, 75.8% of which (45,428 CpGs) were at least partially remethylated by day 15. Out of the 59,920 CpGs hypomethylated by day 8, 21.2% were located in promoters, 50.1% in gene bodies and 28.7% in intergenic regions. In contrast, in T cells only 2 CpGs out of 460,343 CpGs were significantly hypomethylated. This low number is partially due to the higher inter-individual variance as compared to leukemic blasts. Increases in DNA methylation across all patients were very rare, with only 38 CpGs consistently and significantly hypermethylated in blasts and none in T cells. Methylome analysis in granulocytes is currently ongoing. Conclusions: Our mathematical model revealed significant DNA hypomethylation by day 8, with striking remethylation by day 15 from start of decitabine treatment in AML blasts in vivo. Most of the hypomethylated CpGs resided in non-promoter regions. In contrast, T-cells were much less affected, which might be due to the low cell division rate and the fact that they are non-malignant cells. This model will hopefully allow determination whether the effects of decitabine are targeted or random, by including sequential samples from later treatment cycles. Unblinding of the patients' clinical data will reveal potential biomarkers of response to epigenetic therapy. Disclosures Lübbert: Ratiopharm: received study drug valproic acid, received study drug valproic acid Other; Johnson&Johnson: Honoraria, Membership on an entity's Board of Directors or advisory committees, received study drug decitabine Other.


2012 ◽  
Vol 18 (12) ◽  
pp. 2334-2341 ◽  
Author(s):  
Alan R. Harris ◽  
Dorottya Nagy-Szakal ◽  
Natalia Pedersen ◽  
Antone Opekun ◽  
Jiri Bronsky ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document