Dieter Weichenhan's research while affiliated with German Cancer Research Center and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (277)


The promoter mutation paucity as part of the dark matter of the cancer genome
  • Preprint

June 2024

·

12 Reads

Nicholas Allen Baclig Abad

·

Irina Glas

·

·

[...]

·

Cancer is a heterogeneous disease caused by genetic alterations. Computational analysis of cancer genomes led to the expansion of the catalog of driver mutations. While individual high-impact mutations have been discovered also in gene promoters, frequency-based approaches have only characterized a few novel candidates. To investigate the promoter mutation paucity in cancer, we developed the REMIND-Cancer workflow to predict activating promoter mutations in silico, irrespective of their recurrence frequency, and applied it to the PCAWG dataset. We positively validated 7 candidates by luciferase assay including mutations within the promoters of ANKRD53 and MYB. Our analysis indicates that particular mutational signatures and necessary co-alterations constrain the creation and positive selection of functional promoter mutations. We conclude that activating promoter mutations are more frequent in the PCAWG dataset than previously observed, which has potential implications for personalized oncology.

Share

ARID1A regulates DNA repair through chromatin organization and its deficiency triggers DNA damage-mediated anti-tumor immune response

April 2024

·

38 Reads

Nucleic Acids Research

AT-rich interaction domain protein 1A (ARID1A), a SWI/SNF chromatin remodeling complex subunit, is frequently mutated across various cancer entities. Loss of ARID1A leads to DNA repair defects. Here, we show that ARID1A plays epigenetic roles to promote both DNA double-strand breaks (DSBs) repair pathways, non-homologous end-joining (NHEJ) and homologous recombination (HR). ARID1A is accumulated at DSBs after DNA damage and regulates chromatin loops formation by recruiting RAD21 and CTCF to DSBs. Simultaneously, ARID1A facilitates transcription silencing at DSBs in transcriptionally active chromatin by recruiting HDAC1 and RSF1 to control the distribution of activating histone marks, chromatin accessibility, and eviction of RNAPII. ARID1A depletion resulted in enhanced accumulation of micronuclei, activation of cGAS-STING pathway, and an increased expression of immunomodulatory cytokines upon ionizing radiation. Furthermore, low ARID1A expression in cancer patients receiving radiotherapy was associated with higher infiltration of several immune cells. The high mutation rate of ARID1A in various cancer types highlights its clinical relevance as a promising biomarker that correlates with the level of immune regulatory cytokines and estimates the levels of tumor-infiltrating immune cells, which can predict the response to the combination of radio- and immunotherapy.


Proliferative History Is a Novel Driver of Clinical Outcome in Splenic Marginal Zone Lymphoma
  • Preprint
  • File available

January 2024

·

75 Reads

The epiCMIT (epigenetically determined Cumulative MIToses) mitotic clock traces B-cell mitotic history via DNA methylation changes in heterochromatin and H3K27me3-containing chromatin. While high scores correlated with poor outcomes in CLL and MCL, its prognostic significance in SMZL remains unknown. Derived from 142 SMZL cases using DNA methylation microarrays, epiCMIT values were correlated with genomic, transcriptomic, and clinical data. EpiCMIT as a continuous variable was significantly higher in females (p=0.02), patients with IGHV1-2*04 allele usage (p<0001), intermediate IGHV somatic hypermutation load (97-99.9% identity, p=0.04), elevated mutational burden (25 vs. 17 mut/Mb, p=0.001), driver gene mutations [KLF2 (p<0.001), NOTCH2 (p<0.01), TP53 (p=0.01), KMT2D (p<0.001)], and del(7q) (p=0.01). Negative correlation between epiCMIT and telomere length (r=-0.29 p<0.001) supported the association between cumulated proliferation and telomere attrition. While univariate analysis highlighted epiCMIT as robust predictor of shorter treatment-free survival (TFS), multivariate analysis confirmed epiCMIT as an independent marker for shorter TFS. In summary, our matched multi-omic datasets facilitate the clinico-biological characterization of SMZL and introduces epiCMIT as a strong prognostic marker, identifying high-risk patients and predicting reduced treatment-free survival, hence providing a new tool for risk-adapted patient management.

Download

Morphologic and immunohistochemical characteristics of FUS/EWSR1-TFCP2 RMS
a Early lesion (top) showing isomorphic bland spindle cells infiltrating the dermis and subcutaneous fat. Recurrence with histopathologic criteria of malignancy (bottom) 32 months later with significantly increased cellularity, nuclear atypia, and brisk mitoses, composed mainly of spindle cells with intermingled epithelioid/rhabdoid elements. b Early lesion (top) with slightly increased cellularity compared to (a) showing infiltration of the dermis and subcutaneous fat by spindle cells with minimal to low nuclear atypia. Recurrent lesion (bottom) at RMS diagnosis 12 months later with a biphasic pattern consisting of larger areas resembling the spindle cell proliferation of the primary tumor with markedly increased cellularity and nuclear irregularities (part 1) and a second component with a blue cell aspect due to epithelioid/rhabdoid cells with atypical nuclei, coarse chromatin, and brisk mitotic activity (part 2). Stainings were performed once in an accredited pathology laboratory with standardized semi-automated procedures and appropriate controls. H&E, hematoxylin and eosin. Scale bar, 100 µm.
Genetic characteristics of FUS/EWSR1-TFCP2 sarcoma
a ALK and TERT mRNA expression of tumors from all patients enrolled in the MASTER program until November 21, 2018, and all FUS/EWSR1-TFCP2 cases from the MASTER and INFORM studies (indicated in red). TPM, transcripts per million. b Representative ALK transcript variants (see Supplementary Fig. 2c for the remaining cases). The expression levels of ALK exons are indicated by the depth of coverage (pink, regular exons; blue, upstream exons). The expression levels of splice junctions are indicated by the heights of the arcs connecting the exons. Intragenic deletions lead to exon skipping. c Genomic alterations occurring in three or more FUS/EWSR1-TFCP2 sarcoma samples. Copy-number aberrations were filtered for genes listed in the COSMIC Cancer Gene Census (https://cancer.sanger.ac.uk/census), and only homozygous deletions or amplifications with a total copy number above 2.5-fold base ploidy are shown. SNV, single-nucleotide variant; hom., homozygous; SV, structural variant. d Average copy-number profile across all TFCP2-rearranged cases. avg., average. e Dimensionality reduction (t-SNE) based on the expression levels of 792 transcription factors in 282 RNA-seq samples from 277 sarcoma patients, including 14 samples from 12 TFCP2-rearranged cases. f t-SNE using the 6,000 most variable CpG sites (mvCpGs) from 345 DNA methylation profiles of sarcoma samples analyzed in MASTER (n = 343) and INFORM (n = 2). ESS combines samples assigned to methylation class ESS_HG or ESS_LG. CSA combines samples assigned to methylation class CSA_group_A, CSA_group_B, CSA_MES, or CSA_IDH_group_A. In addition, 11 FUS/EWSR1-TFCP2 cases from nine patients were included, which formed two clusters (TFCP2_1 and TFCP2_2), whereas one sample (HD-12) clustered separately. g Spearman correlation of TFCP2_1 (left panel) and TFCP2_2 (right panel) cases with samples from 19 sarcoma entities based on the same 6000 mvCpGs. Entities are sorted by decreasing correlation from top to bottom. The color code is the same as in f. TFCP2 FUS/EWSR1-TFCP2 sarcoma, RMS_ALV alveolar RMS, RMS_MYOD1 MYOD1-mutant spindle cell/sclerosing RMS, RMS_EMB embryonal RMS, MPNST malignant peripheral nerve sheath tumor, WDLS_DDLS well-differentiated and dedifferentiated liposarcoma, SFT solitary fibrous tumor, CCS clear cell sarcoma, ES epithelioid sarcoma, USARC undifferentiated sarcoma, CHORD chordoma, ASPS alveolar soft part sarcoma, SEF sclerosing epithelioid sarcoma, GIST gastrointestinal stromal tumor, AS angiosarcoma, CSA chondrosarcoma, OS_HG osteosarcoma high-grade, DSRCT desmoplastic small round cell tumor, ESS endometrial stromal sarcoma, EWING Ewing sarcoma, MLS myxoid liposarcoma, SYSA synovial sarcoma.
Functional and structural characteristics of ALK alterations associated with FUS/EWSR1-TFCP2 RMS
a Colony formation of MCF10A cells stably transduced with ALK alterations or EV. Mean ± SEM (n = 3 independent experiments). b Anchorage-independent growth in soft agar of MCF10A cells stably transduced with ALK alterations or EV. Mean ± SEM (n = 3 independent experiments). c Tumor growth in NOD-SCID mice of MCF10A cells stably transduced with ALK alterations or EV. Shown is the mean volume ± SEM of six tumors per cell line until the first mouse had to be sacrificed in one group due to reaching the maximum allowed tumor length. d Sensitivity of MCF10A cells stably transduced with the indicated ALK variants or EV to crizotinib, ceritinib, or alectinib after 72 h in the absence of EGF. Drug concentrations (conc.) are shown at the top of the heatmap. Mean cell viability of drug-treated cells relative to the respective DMSO control (n = 2 independent experiments). e Sensitivity of freshly isolated and singularized cells from tumor sample TFCP2-HD-4 to the ALK inhibitors crizotinib, ceritinib, or alectinib. Vertical dotted lines represent the Cmax of each compound. Mean ± SEM (n = 4 technical replicates). f Domain architectures of ALK variants. Pairings between cysteines in each variant are indicated by red lines. SP signal peptide, MAM meprin A-5 protein, and receptor protein-tyrosine phosphatase mu domain, LDL low-density lipoprotein receptor class A, EGF epidermal growth factor-like domain, TM transmembrane helix, Pkinase_tyr tyrosine protein kinase domain. Statistical significance was assessed by a one-tailed unpaired t-test. ns not significant. Source data for a–e are provided in the Source Data file.
Effects of TFCP2 fusions on myogenic differentiation
a Stages of muscle cell differentiation from myoblasts to myotubes and corresponding expression pattern of MYOD and MYOG transcription factors and myosin heavy chain (MHC). The image was created with BioRender.com and adapted from Bentzinger et al.⁶⁴. b Immunofluorescence images of LHCN-M2 cells transduced with EV or FUS-TFCP2 and cultured in differentiation medium for six days. Green, MyoHC; blue, DAPI (nuclei). Representative images of three independent experiments with similar results are shown. Scale bar, 1 mm. c Fusion index of LHCN-M2 cells transduced with EV or FUS-TFCP2 and cultured in differentiation medium for five days. Mean ± SEM (n = 3 independent experiments). d Relative MYOD and MYOG mRNA expression of LHCN-M2 cells transduced with EV or FUS-TFCP2 and cultured in differentiation medium for six days. Mean ± SEM (n = 3 independent experiments). e Phase-contrast images of LHCN-M2 cells transduced with EV, FUS, TFCP2, or FUS-TFCP2 and cultured in differentiation medium for ten days. Representative images of six independent experiments with similar results are shown. Scale bar, 1 mm. Source data for c and d are provided in the Source Data file.
Transcriptional effects of TFCP2 fusions
a Relative FUS-TFCP2 and ALK mRNA expression in MCF10A cells stably transduced with FUS-TFCP2. Mean ± SEM (n = 3 independently transduced cell lines). Source data are provided in the Source Data file. b Number of genes significantly deregulated in MCF10A and SCP-1 cells transduced with FUS-TFCP2 or EWSR1-TFCP2 versus cells transduced with EV (log2(fold-change) >1.0 or <–1.0), as determined by RNA-seq. c Genes significantly deregulated in MCF10A and SCP-1 cells transduced with FUS-TFCP2 or EWSR1-TFCP2 versus cells transduced with EV (log2(fold-change) >1.0 or <–1.0 in at least three cell lines), as determined by RNA-seq. d Expression of genes from c in FUS/EWSR1-TFCP2-positive sarcoma samples, indicated as percentiles of expression across the entire MASTER cohort. e, f Genome browser images of ALK (e) and TERT (f) showing enrichment peaks obtained by ACT-seq with an anti-HA antibody (Cell Signaling) in MCF10A cells stably expressing EV, HA-TFCP2, or HA-FUS-TFCP2. g Genome browser image showing aberrant transcription originating from the second intron of TERT in patient TFCP2-HD-1. The first two exons were not transcribed. Instead, multiple novel intronic transcription start sites (red alignments) and antisense transcription (blue reads) were found around putative TFCP2 binding sites (bottom) detected by HOMER using the known Tcfcp2l1 binding motif. h Domain structure of full-length TERT (top) and the TERT variant predicted to be translated from mRNA lacking exons 1 and 2 (bottom). The domain structure was adapted from Chan et al.⁶⁵. CTE C-terminal extension, RT reverse transcriptase, TEN TERT-essential N-terminal, TRBD telomerase RNA-binding domain. i Western blot with tumor tissue from five patients and HeLa cells with an antibody binding to the C-terminus of TERT. Protein masses in kDa are shown on the left. Uncropped blots are provided in the Source Data file.

+1

Multi-omic and functional analysis for classification and treatment of sarcomas with FUS-TFCP2 or EWSR1-TFCP2 fusions

January 2024

·

141 Reads

Nature Communications

Linking clinical multi-omics with mechanistic studies may improve the understanding of rare cancers. We leverage two precision oncology programs to investigate rhabdomyosarcoma with FUS/EWSR1-TFCP2 fusions, an orphan malignancy without effective therapies. All tumors exhibit outlier ALK expression, partly accompanied by intragenic deletions and aberrant splicing resulting in ALK variants that are oncogenic and sensitive to ALK inhibitors. Additionally, recurrent CKDN2A/MTAP co-deletions provide a rationale for PRMT5-targeted therapies. Functional studies show that FUS-TFCP2 blocks myogenic differentiation, induces transcription of ALK and truncated TERT, and inhibits DNA repair. Unlike other fusion-driven sarcomas, TFCP2-rearranged tumors exhibit genomic instability and signs of defective homologous recombination. DNA methylation profiling demonstrates a close relationship with undifferentiated sarcomas. In two patients, sarcoma was preceded by benign lesions carrying FUS-TFCP2, indicating stepwise sarcomagenesis. This study illustrates the potential of linking precision oncology with preclinical research to gain insight into the classification, pathogenesis, and therapeutic vulnerabilities of rare cancers.


The Proliferative History Index, Epicmit, Derived from Genome-Wide Epigenomic Profiling, Is a Key Driver of Clinical Survival in Splenic Marginal Zone Lymphoma

November 2023

·

22 Reads

Blood

The epiCMIT (epigenetically-determined Cumulative MIToses) mitotic clock (Durran-Ferrer, 2020) tracks DNA hypo- and hyper-methylation changes in heterochromatin and H3K27me3-containing chromatin, respectively, providing a comprehensive record of B cell mitotic history (pre- and post-malignant transformation). Higher epiCMIT levels correlate with poor survival in CLL and MCL, suggesting that a greater mitotic history predicts future proliferative capacity. However, no studies have investigated epiCMIT in splenic marginal zone lymphoma (SMZL). We studied 142 SMZL cases, a rare lymphoid malignancy with specific genetic characteristics [del(7q), KLF2and NOTCH2 mutations, biased use of IGHV1-2*04]. After calculating epiCMIT scores from 450/850K Illumina arrays we compared these data with gene mutations from targeted resequencing and WGS, copy number variations, telomere length, RNA-Seq, miRNA-Seq data, and clinical outcome data. By defining epiCMIT-hyper [mean: 0.6, range: 0.29-0.91] and -hypo [mean: 0.71, range: 0.39-0.90] values in our cohort, we demonstrate a strong positive correlation between hyper- and hypo- scores, as previously observed in MCL and CLL (Durran-Ferrer, 2020). The highest score obtained for epiCMIT-hyper or -hypo per sample was then selected as the epiCMIT score per patient [mean: 0.72, range: 0.39-0.91]. We identified significant correlations between epiCMIT and key clinico-biological characteristics, including a significant negative correlation between epiCMIT and telomere length (R 2=0.67 p<0.001) supporting cell proliferation and concomitant telomere attrition. High epiCMIT scores were associated with female gender (p=0.02), IGHV1-2*04 alleles (p<0.001) with intermediate levels of IGHV somatic hypermutation (SHM) load (97-99.9%, p=0.04), driver somatic gene mutations, including those in KLF2 (p<0.001), NOTCH2 (p<0.01), TP53 (p=0.01) and KMT2D (p<0.001) and deletion of 7q (p=0.01) ( Fig1). Using previously defined classification systems (Arribas, 2015 and Bonfiglio, 2022), we show enrichment of high-risk subgroups [Arribas- high global methylation subgroup (High-M, p<0.001) and Bonfiglio- NFKB, NOTCH and KLF2 genetic subgroup (NNK, p=0.01)] in cases with high epiCMIT. In a sub-cohort of 42 SMZL cases, with a spectrum of epiCMIT values and WGS/ (mi)RNA-Seq data available, we demonstrated 1) a significantly higher genome-wide somatic mutational burden in patients with high epiCMIT-total scores (25 v 17mut/Mb, p=0.001), and 2), correlations between epiCMIT and specific gene/miRNA expression associated with cellular survival and proliferation ( CARD11 [R 2=0.6, p<0.01], MAP2K1 [R 2=0.61, p<0.01], miRNA-155 [R 2=0.58, p=0.018]), apoptosis ( BCOR [R 2=0.61, p<0.01]), and epigenetic regulation ( EZH2 [R 2=0.58, p<0.01]). We then conducted univariate Cox Proportional Hazards analysis to investigate the prognostic significance of clinical and molecular features on time to first treatment (TTFT; most prevalent treatments: splenectomy, n=51; rituximab, n=20) and overall survival (OS). The epiCMIT score and epiCMIT-hyper values showed the highest hazard ratios (HR) for TTFT (HR=26.6, p=0.001) and OS (HR=9.45, p=0.043) respectively ( Fig2). Other factors associated with shorter TTFT included TP53 mutation, 3q gain, IGHV1-2*04 usage, High-M, short telomeres and female gender. Shorter OS was significantly (p<0.05) linked to mutations in key genes (including TP53 and NOTCH2), genomic complexity and del(7q). To avoid redundancy and collinearity, only the most significant epiCMIT metric was included in a multivariate Cox Proportional Hazards model (backwards selection, 94 patients/63 events), and epiCMIT-total emerged as an independent marker for shorter TTFT (HR=44.4, p=0.004), together with gain of chromosome 3q (HR=2.6, p=0.002). Kaplan-Meier analysis further confirmed the importance of epiCMIT-total, with higher scores (>median: 0.73) associated with significantly shorter TTFT (median 23 vs 3.5 months) and higher mortality (p=0.004). To conclude, we demonstrate the potential clinical utility of epiCMIT score in SMZL patients, identifying that SMZL patients with high epiCMIT harbour specific genetic characteristics, and exhibit reduced treatment-free survival. EpiCMIT could be valuable in clinical practice, helping to identify those patients destined to progress and require closer monitoring.


Predictive value of DNA methylation patterns in AML patients treated with an azacytidine containing induction regimen

October 2023

·

43 Reads

Clinical Epigenetics

Background Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis. Dysregulation of the epigenetic machinery is a significant contributor to disease development. Some AML patients benefit from treatment with hypomethylating agents (HMAs), but no predictive biomarkers for therapy response exist. Here, we investigated whether unbiased genome-wide assessment of pre-treatment DNA-methylation profiles in AML bone marrow blasts can help to identify patients who will achieve a remission after an azacytidine-containing induction regimen. Results A total of n = 155 patients with newly diagnosed AML treated in the AMLSG 12-09 trial were randomly assigned to a screening and a refinement and validation cohort. The cohorts were divided according to azacytidine-containing induction regimens and response status. Methylation status was assessed for 664,227 500-bp-regions using methyl-CpG immunoprecipitation-seq, resulting in 1755 differentially methylated regions (DMRs). Top regions were distilled and included genes such as WNT10A and GATA3. 80% of regions identified as a hit were represented on HumanMethlyation 450k Bead Chips. Quantitative methylation analysis confirmed 90% of these regions (36 of 40 DMRs). A classifier was trained using penalized logistic regression and fivefold cross validation containing 17 CpGs. Validation based on mass spectra generated by MALDI-TOF failed (AUC 0.59). However, discriminative ability was maintained by adding neighboring CpGs. A recomposed classifier with 12 CpGs resulted in an AUC of 0.77. When evaluated in the non-azacytidine containing group, the AUC was 0.76. Conclusions Our analysis evaluated the value of a whole genome methyl-CpG screening assay for the identification of informative methylation changes. We also compared the informative content and discriminatory power of regions and single CpGs for predicting response to therapy. The relevance of the identified DMRs is supported by their association with key regulatory processes of oncogenic transformation and support the idea of relevant DMRs being enriched at distinct loci rather than evenly distribution across the genome. Predictive response to therapy could be established but lacked specificity for treatment with azacytidine. Our results suggest that a predictive epigenotype carries its methylation information at a complex, genome-wide level, that is confined to regions, rather than to single CpGs. With increasing application of combinatorial regimens, response prediction may become even more complicated.


Fig. 2. MNX1 expression in pediatric AML with t(7;12)(q36;p13) translocation. MNX1 expression in two different pediatric AML cohorts: a, Balgobind et al. 2011 18 , 237 samples profiled with Affymetrix arrays. The mean expression level is shown with a dashed line, and the mean plus three standard deviations is shown with a horizontal line. b, TARGET-AML 15 , 1319 samples profiled with RNAseq (transcripts per million, TPM). A cut-off of 0.5 TPM was used. Samples with cytogenetically detected t(7;12)(q36;p13) translocation are shown in green and other samples in grey.
Fig. 3. MNX1 protein expression and chromatin interaction of the MNX1 gene with the ETV6 region in ChiPSC22 t(7;12) HSPCs. a, Western blot with an MNX1 antibody (left) and iPSC (blue) and HSPC (red) protein extracts from ChiPSC22 WT and ChiPSC22 t(7;12) sublines 14D7, 23G8 and 24C7. The MNX1 protein (asterisk) is only detected in HSPCs of ChiPSC22 t(7;12) sublines 14D7, 23G8 and 24C7. The common band at about 120 kD results from an unknown protein cross-reacting with the MNX1 antibody. To demonstrate loading of equal protein amounts, the unstripped blot was reincubated with a horseradish-peroxidase coupled antibody against β-actin (right). b, Circular chromosome conformation capture (4C) indicates specific interaction between the MNX1 (viewpoint about 3 kb upstream of MNX1) and the ETV6 region in HSPCs of ChiPSC22 t(7;12) sublines 14D7 and 24C7. The range, 0-3522, of read numbers which are indicated by vertical bars was given by sample 24C7 HSPC and set to same for all samples. c, Chromatin interactions analyzed by Hi-C seq in the genomic region flanking the translocation breakpoint in the ChiPSC22 t(7;12) subline 24C7, either as iPSCs (left) or HSPCs (right). Below, gene locations and peaks of H3K27ac, H3K4me1 and open chromatin are shown to indicate locations of putative enhancers.
Fig. 4. Enhancer mark profiles 1 Mbp distal to the breakpoint (BP) position in ETV6 of ChiPSC22 t(7;12) sublines 14D7, 23G8 and 24C7 and of ChiPSC22 WT . a, HSPC-specific H3K27ac and b, HSPC-specific H3K4me1 profiles. Publicly available respective profiles of CD34+ and MOLM-1 (also for enhancer mark P300) are shown as well. Relevant common peak positions in a and b are highlighted by a gray shading.
Fig. 5 Open chromatin and enhancer mark profiles in the ETV6 region of patient and cell line samples. a, Open chromatin profiles of AML patients AML-T1 and -T2 and of ChiPSC22 t(7;12) sublines 14D7, 23G8 and 24C7 and of ChiPSC22 WT HSPCs 1 MB distal to the breakpoint (BP) in ETV6. b, High resolution open chromatin profiles in the enhancer region 1 of AML-T1, AML-T2 and of iPSCs and HSPCs of ChiPSC22 t(7;12) and ChiPSC22 WT . c and d, High resolution H3K27ac and H3K4me1, respectively, profiles in the enhancer region 1 of iPSCs and HSPCs of ChiPSC22 t(7;12) and ChiPSC22 WT . A publicly available P300 profile of MOLM-1 is shown as well. Relevant common peak positions are highlighted by a gray shading. Enhancer region 1 is resolved into enhancer 1a, active in iPSCs, and enhancer 1b, active in HSPCs.
Fig. 6 Molecular validation of enhancer-promoter interaction in ChiPSC22 t(7;12) upon differentiation. a, Scheme of experiments performed to validate the interaction between the MNX1 promoter and enhancers distal to the breakpoint including deleting the enhancer region and spatial proximity probing. b, Gene expression in ChiPSC22 WT (n=3), ChiPSC22 t(7;12) (n=6, from 3 independent cell lines) and ChiPSC22 t(7;12)ΔEn (n=5, from 3 independent cell lines) measured via qRT-PCR and shown as 2 −ΔCt vs. GUSB as endogenous reference. Unpaired, two-tailed t-test with *P < 0.05 and ***P < 0.0001. c, Increased proximity between MNX1 and ETV6 in ChiPSC22 t(7;12) derived HSPCs compared to iPSCs. Left: 2-color FISH targeting a 5 kb region at MNX1 promoter and putative enhancer 1 in ETV6 in iPSCs and HSPCs. Representative single planes are depicted. Note that the iPSC representative image was scaled down. Scale bars, 5 µm. Right: 3D distances between MNX1 and ETV6 upon differentiation. Distances under 3.5 µm are displayed. Black horizontal lines within boxes represent median values. The limits of the boxes indicate the upper and lower quartiles. iPSC: n = 64 , HSPC: n = 90, across 3 independent replicates. *P < 0.05, Wilcoxon rank sum test.
Altered enhancer-promoter interaction leads to MNX1 expression in pediatric acute myeloid leukemia with t(7;12)(q36;p13)

September 2023

·

157 Reads

·

1 Citation

Acute myeloid leukemia (AML) with the t(7;12)(q36;p13) translocation occurs only in very young children and has a poor clinical outcome. The expected oncofusion between breakpoint partners ( MNX1 and ETV6 ) has only been reported in a subset of cases. However, a universal feature is the strong transcript and protein expression of MNX1, a homeobox transcription factor that is normally not expressed in hematopoietic cells. Here, we map the translocation breakpoints on chromosomes 7 and 12 in affected patients to a region proximal to MNX1 and either introns 1 or 2 of ETV6 . The frequency of MNX1 overexpression in pediatric AML (n=1556, own and published data) is 2.4% and occurs predominantly in t(7;12)(q36;p13) AML. Chromatin interaction assays in a t(7;12)(q36;p13) iPSC cell line model unravel an enhancer-hijacking event that explains MNX1 overexpression in hematopoietic cells. Our data suggest that enhancer-hijacking is a more common and overlooked mechanism for structural rearrangement-mediated gene activation in AML. Key points Expression analysis of over 1500 pediatric AML samples demonstrates MNX1 expression as a universal feature of t(7;12)(q36;p13) AML as well as in rare cases without t(7;12)(q36;p13) MNX1 is activated by an enhancer-hijacking event in t(7;12)(q36;p13) AML and not, as previously postulated, by the creation of a MNX1 :: ETV6 oncofusion gene.


S120: MNX1-ACTIVATING ENHANCER HIJACKING EVENTS IN ACUTE MYELOID LEUKEMIA WITH DELETIONS ON CHROMOSOME 7Q

August 2023

·

53 Reads

HemaSphere

HemaSphere




Citations (53)


... Indeed, the translocated intron 1 of ETV6 contains numerous enhancers and regulatory elements that may drive MNX1 overexpression, mimicking the molecular mechanism described for canonical t(7;12) cases [11]. Of note, a recent study points to an enhancer-hijacking event activating the MNX1 promoter from the ETV6 locus as an explanation for the MNX1 overexpression in this AML subtype [15]. Overall, these observations support that MNX1, rather than NOM1, is most likely the driver event of the disease, akin to other t(7;12) leukemias [13]. ...

Reference:

Backtracking NOM1::ETV6 fusion to neonatal pathogenesis of t(7;12) (q36;p13) infant AML
Altered enhancer-promoter interaction leads to MNX1 expression in pediatric acute myeloid leukemia with t(7;12)(q36;p13)

... MNX1 overexpression does not lead to leukemic transformation of cord blood (CB) cells or adult mouse bone marrow (BM) cells, but impedes erythroid differentiation and encourages cellular senescence [12]. However, MNX1 overexpression in fetal liver cells, but not in adult BM cells, leads to leukemic transformation in a retroviral mouse model [13]. MNX1::ETV6, independently of MNX1 overexpression, does not confer self-renewal capacity or leukemogenic potential in a model involving transduced fetal liver cells. ...

Aberrant MNX1 expression associated with t(7;12)(q36;p13) pediatric acute myeloid leukemia induces the disease through altering histone methylation

Haematologica

... These changes can alter the expression/silencing of the target genes, increasing the risk of developing COPD. The DNA methylation patterns of human lung fibroblasts of different COPD stages were recently analyzed [46]. The high-resolution multi-omic analysis demonstrated that significant changes, particularly in genome regulatory regions, occur at an early stage. ...

High-resolution transcriptomic and epigenetic profiling identifies novel regulators of COPD

The EMBO Journal

... Cell lines continue to be a vital resource for functional studies. Several well-established cancer cell lines are highly rearranged, with underreported genomic complexity Aganezov et al, 2020;Weichenhan et al, 2023). Despite its frequent use for functional and mechanistic studies in leukemia research (Oshima et al, 2020;Diedrich et al, 2021;Cousins et al, 2022;Leo et al, 2022;Wray et al, 2022), a comprehensive analysis of the complex REH genome has never been undertaken. ...

Translocation t(6;7) in AML-M4 cell line GDM-1 results in MNX1 activation through enhancer-hijacking

Leukemia

... DNA methylation is involved and further, enforced influence in the occurrence and development of cardiac hypertrophy [13,14], which was demonstrated by Tong-Tong Wu and his colleagues in the heart tissues of HF mice [15]. Meder et al. analyzed plasma-treated hiPSC-CMs and cardiac biopsies using array-based DNA methylation analysis and revealed prevalent alterations in cardiac DNA methylation; they further revealed that hypomethylation of the ATG promoter could serve as a diagnostic marker of HF [16]. Cameron et al. found large differences in DNA methylation at over 16,400 CpG methylation sites, particularly in the canonical signaling pathway of Cardiac Hypertrophy in neonates [17]. ...

Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure

Basic Research in Cardiology

... [4][5][6] It has become evident that a considerable part of the increase in asthma prevalence worldwide is due to maternal smoking in pregnancy or due to early-life exposure of children to ETS acting through epigenetic transgenerational effects. [14][15][16][17] The recently published SR of 67 longitudinal studies proved with moderate certainty evidence that prenatal ETS increases the risk of recurrent wheeze and may increase the risk of new-onset asthma and low lung function. 12 Postnatal ETS increases the risk of new-onset asthma and recurrent wheezing and may impact lung function. ...

Global hypomethylation in childhood asthma identified by genome‐wide DNA‐methylation sequencing preferentially affects enhancer regions

... The tumor homologous recombination deficiency (HRD) genomic instability score (GIS) calculated by combining three factors, LOH, telomeric allelic imbalance, and large-scale state transitions, was of 15, below the validated threshold of 42 in the context of ovarian cancers [19]. Platinum drug treatment mutational signature SBS35 according to the COSMIC classification [20] contributed to 22% of somatic variants while SBS5 signature of unknown etiology and SBS1 spontaneous deamination of 5-methylcytosine signature contributed to 63% and 15% of somatic variants respectively. No SBS3 nor SBS8 homologous recombination deficiency signature was observed. ...

Author Correction: The repertoire of mutational signatures in human cancer

Nature

... Human malignancies are characterized by chromosome CNVs, which include genes that are in hundreds or thousands [3,17]. According to a growing amount of evidence, CNVs are recurring in several cancer types, with some of them having a probability of appearing in the early tumorigenesis stages, demonstrating that some cancer-associated CNVs may serve as human cancers drivers [30,31]. CNAs can speed up tumor growth by changing the gene expression levels whose location is at regions of the impacted genomic [32]. ...

Author Correction: The evolutionary history of 2,658 cancers

Nature

... Structural variations (SVs) are large-size genetic variations in the human genome, and include insertion, deletion, duplication, inversion, and translocation. SVs have been associated with different traits and with various diseases, including breast cancer [4][5][6]. They contribute to gene fusion, oncogene amplification, tumor suppressor gene deletion and other complex alterations leading to evolution of the cancer genome. ...

Author Correction: Patterns of somatic structural variation in human cancer genomes

Nature

... P-values were computed using t-statistics from linear regression in the R package Matrix eQTL [29]. We used a two-step multiple-testing correction procedure, as described in [30]. First, for each gene, we correct for the number of variants tested using Bonferroni correction. ...

Author Correction: Genomic basis for RNA alterations in cancer

Nature