Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Syst Biol ; 18(9): e11073, 2022 09.
Article in English | MEDLINE | ID: mdl-36121124

ABSTRACT

Keratinocyte cancers (KC) are the most prevalent malignancies in fair-skinned populations, posing a significant medical and economic burden to health systems. KC originate in the epidermis and mainly comprise basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (cSCC). Here, we combined single-cell multi-omics, transcriptomics, and methylomics to investigate the epigenomic dynamics during epidermal differentiation. We identified ~3,800 differentially accessible regions between undifferentiated and differentiated keratinocytes, corresponding to regulatory regions associated with key transcription factors. DNA methylation at these regions defined AK/cSCC subtypes with epidermal stem cell- or keratinocyte-like features. Using cell-type deconvolution tools and integration of bulk and single-cell methylomes, we demonstrate that these subclasses are consistent with distinct cells-of-origin. Further characterization of the phenotypic traits of the subclasses and the study of additional unstratified KC entities uncovered distinct clinical features for the subclasses, linking invasive and metastatic KC cases with undifferentiated cells-of-origin. Our study provides a thorough characterization of the epigenomic dynamics underlying human keratinocyte differentiation and uncovers novel links between KC cells-of-origin and their prognosis.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Epigenomics , Humans , Keratinocytes/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors
2.
Stem Cells ; 39(9): 1270-1284, 2021 09.
Article in English | MEDLINE | ID: mdl-34013984

ABSTRACT

Acute myeloid leukemia (AML) is characterized by an expansion of leukemic cells and a simultaneous reduction of normal hematopoietic precursors in the bone marrow (BM) resulting in hematopoietic insufficiency, but the underlying mechanisms are poorly understood in humans. Assuming that leukemic cells functionally inhibit healthy CD34+ hematopoietic stem and progenitor cells (HSPC) via humoral factors, we exposed healthy BM-derived CD34+ HSPC to cell-free supernatants derived from AML cell lines as well as from 24 newly diagnosed AML patients. Exposure to AML-derived supernatants significantly inhibited proliferation, cell cycling, colony formation, and differentiation of healthy CD34+ HSPC. RNA sequencing of healthy CD34+ HSPC after exposure to leukemic conditions revealed a specific signature of genes related to proliferation, cell-cycle regulation, and differentiation, thereby reflecting their functional inhibition on a molecular level. Experiments with paired patient samples showed that these inhibitory effects are markedly related to the immunomagnetically enriched CD34+ leukemic cell population. Using PCR, ELISA, and RNA sequencing, we detected overexpression of TGFß1 in leukemic cells on the transcriptional and protein level and, correspondingly, a molecular signature related to TGFß1 signaling in healthy CD34+ HSPC. This inhibitory effect of TGFß1 on healthy hematopoiesis was functionally corrobated and could be pharmacologically reverted by SD208, an inhibitor of TGFß receptor 1 signaling. Overall, these data indicate that leukemic cells induce functional inhibition of healthy CD34+ HSPC, at least in part, through TGFß1, suggesting that blockage of this pathway may improve hematopoiesis in AML.


Subject(s)
Hematopoietic Stem Cells , Leukemia, Myeloid, Acute , Antigens, CD34/metabolism , Bone Marrow/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics
3.
PLoS Pathog ; 14(11): e1007420, 2018 11.
Article in English | MEDLINE | ID: mdl-30485383

ABSTRACT

It is currently believed that type I and III interferons (IFNs) have redundant functions. However, the preferential distribution of type III IFN receptor on epithelial cells suggests functional differences at epithelial surfaces. Here, using human intestinal epithelial cells we could show that although both type I and type III IFNs confer an antiviral state to the cells, they do so with distinct kinetics. Type I IFN signaling is characterized by an acute strong induction of interferon stimulated genes (ISGs) and confers fast antiviral protection. On the contrary, the slow acting type III IFN mediated antiviral protection is characterized by a weaker induction of ISGs in a delayed manner compared to type I IFN. Moreover, while transcript profiling revealed that both IFNs induced a similar set of ISGs, their temporal expression strictly depended on the IFNs, thereby leading to unique antiviral environments. Using a combination of data-driven mathematical modeling and experimental validation, we addressed the molecular reason for this differential kinetic of ISG expression. We could demonstrate that these kinetic differences are intrinsic to each signaling pathway and not due to different expression levels of the corresponding IFN receptors. We report that type III IFN is specifically tailored to act in specific cell types not only due to the restriction of its receptor but also by providing target cells with a distinct antiviral environment compared to type I IFN. We propose that this specific environment is key at surfaces that are often challenged with the extracellular environment.


Subject(s)
Interferon Type I/genetics , Interferons/genetics , Antiviral Agents/pharmacology , Cell Line , Epithelial Cells/metabolism , Humans , Interferon Type I/metabolism , Interferons/metabolism , Interleukins/metabolism , Intestinal Mucosa/metabolism , Receptors, Interferon/genetics , Signal Transduction/drug effects , Interferon Lambda
4.
Methods ; 156: 121-127, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30366099

ABSTRACT

RNA cytosine-5 methylation (m5C) has emerged as a key epitranscriptomic mark, which fulfills multiple roles in structural modulation, stress signaling and the regulation of protein translation. Bisulfite sequencing is currently the most accurate and reliable method to detect m5C marks at nucleotide resolution. Targeted bisulfite sequencing allows m5C detection at single base resolution, by combining the use of tailored primers with bisulfite treatment. A number of computational tools currently exist to analyse m5C marks in DNA bisulfite sequencing. However, these methods are not directly applicable to the analysis of RNA m5C marks, because DNA analysis focuses on CpG methylation, and because artifactual unconversion and misamplification in RNA can obscure actual methylation signals. We describe a pipeline designed specifically for RNA cytosine-5 methylation analysis in targeted bisulfite sequencing experiments. The pipeline is directly applicable to Illumina MiSeq (or equivalent) sequencing datasets using a web interface (https://bisamp.dkfz.de), and is defined by optimized mapping parameters and the application of tailored filters for the removal of artifacts. We provide examples for the application of this pipeline in the unambiguous detection of m5C marks in tRNAs from mouse embryonic stem cells and neuron-differentiated stem cells as well as in 28S rRNA from human fibroblasts. Finally, we also discuss the adaptability of BisAMP to the analysis of DNA methylation. Our pipeline provides an accurate, fast and user-friendly framework for the analysis of cytosine-5 methylation in amplicons from bisulfite-treated RNA.


Subject(s)
5-Methylcytosine/analysis , Cytosine/metabolism , High-Throughput Nucleotide Sequencing/methods , RNA, Ribosomal, 28S/chemistry , RNA, Transfer/chemistry , Sulfites/chemistry , Transcriptome , 5-Methylcytosine/metabolism , Animals , Cell Differentiation , DNA/genetics , DNA/metabolism , DNA Methylation , Datasets as Topic , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Internet , Methylation , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Nucleic Acid Conformation , RNA, Ribosomal, 28S/genetics , RNA, Ribosomal, 28S/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Software
5.
Int J Cancer ; 144(3): 569-581, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30252132

ABSTRACT

Expression of the epidermal growth factor ligands amphiregulin (AREG) and epiregulin (EREG) is positively correlated with a response to EGFR-targeted therapies in colorectal cancer. Gene-body methylation sites, which show a strong inverse correlation with AREG and EREG gene expression, were identified in cell lines using targeted 454 FLX-bisulfite sequencing and SIRPH analyses for AREG/EREG promoters and intragenic CpGs. Upon treatment of colorectal cancer cells with 5-aza-2'-desoxycytidine, methylation decreases at specific intragenic CpGs accompanied by upregulation of AREG and EREG gene expression. The same AREG gene-body methylation was also found in human colorectal cancer samples and is independent of KRAS and NRAS mutations. Methylation is specifically decreased in the tumor epithelial compartment as compared to stromal tissue and normal epithelium. Investigation of a promoter/enhancer function of the AREG exon 2 region revealed a potential promoter function in reverse orientation. Retrospective comparison of the predictive power of AREG gene-body methylation versus AREG gene expression using samples from colorectal cancer patients treated with anti-EGFR inhibitors with complete clinical follow-up revealed that AREG expression is superior to AREG gene methylation. AREG and EREG genes undergo a complex regulation involving both intragenic methylation and promoter-dependent control.


Subject(s)
Amphiregulin/genetics , Colorectal Neoplasms/genetics , Epiregulin/genetics , Amphiregulin/biosynthesis , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Epigenesis, Genetic , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression , HCT116 Cells , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Retrospective Studies , Stromal Cells/metabolism , Stromal Cells/pathology
6.
Haematologica ; 103(9): 1462-1471, 2018 09.
Article in English | MEDLINE | ID: mdl-29773599

ABSTRACT

Mesenchymal stromal cells are involved in the pathogenesis of myelodysplastic syndromes and acute myeloid leukemia, but the underlying mechanisms are incompletely understood. To further characterize the pathological phenotype we performed RNA sequencing of mesenchymal stromal cells from patients with myelodysplastic syndromes and acute myeloid leukemia and found a specific molecular signature of genes commonly deregulated in these disorders. Pathway analysis showed a strong enrichment of genes related to osteogenesis, senescence, inflammation and inhibitory cytokines, thereby reflecting the structural and functional deficits of mesenchymal stromal cells in myelodysplastic syndromes and acute myeloid leukemia on a molecular level. Further analysis identified transforming growth factor ß1 as the most probable extrinsic trigger factor for this altered gene expression. Following exposure to transforming growth factor ß1, healthy mesenchymal stromal cells developed functional deficits and adopted a phenotype reminiscent of that observed in patient-derived stromal cells. These suppressive effects of transforming growth factor ß1 on stromal cell functionality were abrogated by SD-208, an established inhibitor of transforming growth factor ß receptor signaling. Blockade of transforming growth factor ß signaling by SD-208 also restored the osteogenic differentiation capacity of patient-derived stromal cells, thus confirming the role of transforming growth factor ß1 in the bone marrow microenvironment of patients with myelodysplastic syndromes and acute myeloid leukemia. Our findings establish transforming growth factor ß1 as a relevant trigger causing functional inhibition of mesenchymal stromal cells in myelodysplastic syndromes and acute myeloid leukemia and identify SD-208 as a candidate to revert these effects.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mesenchymal Stem Cells/metabolism , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Transforming Growth Factor beta1/genetics , Adult , Aged , Aged, 80 and over , Biomarkers , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/pathology , Male , Mesenchymal Stem Cells/drug effects , Middle Aged , Myelodysplastic Syndromes/pathology , Osteogenesis/drug effects , Osteogenesis/genetics , Phenotype , Pteridines/pharmacology , Sequence Analysis, RNA , Signal Transduction , Transforming Growth Factor beta1/metabolism
7.
Blood Adv ; 8(10): 2575-2588, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38241490

ABSTRACT

ABSTRACT: The hallmark of multiple myeloma (MM) is a clonal plasma cell infiltration in the bone marrow accompanied by myelosuppression and osteolysis. Premalignant stages such as monoclonal gammopathy of undetermined significance (MGUS) and asymptomatic stages such as smoldering myeloma (SMM) can progress to MM. Mesenchymal stromal cells (MSCs) are an integral component of the bone marrow microenvironment and play an important role in osteoblast differentiation and hematopoietic support. Although stromal alterations have been reported in MM contributing to hematopoietic insufficiency and osteolysis, it is not clear whether alterations in MSC already occur in MGUS or SMM. In this study, we analyzed MSCs from MGUS, SMM, and MM regarding their properties and functionality and performed messenger RNA sequencing to find underlying molecular signatures in different disease stages. A high number of senescent cells and a reduced osteogenic differentiation capacity and hematopoietic support were already present in MGUS MSC. As shown by RNA sequencing, there was a broad spectrum of differentially expressed genes including genes of the BMP/TGF-signaling pathway, detected already in MGUS and that clearly increases in patients with SMM and MM. Our data may help to block these signaling pathways in the future to hinder progression to MM.


Subject(s)
Mesenchymal Stem Cells , Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Smoldering Multiple Myeloma , Humans , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Mesenchymal Stem Cells/metabolism , Cell Differentiation , Male , Female , Aged
8.
Cancers (Basel) ; 16(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38893194

ABSTRACT

Myeloid and lymphoid neoplasms share the characteristics of potential bone marrow infiltration as a primary or secondary effect, which readily leads to hematopoietic insufficiency. The mechanisms by which clonal malignant cells inhibit normal hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM) have not been unraveled so far. Given the pivotal role of mesenchymal stromal cells (MSCs) in the regulation of hematopoiesis in the BM niche it is assumed that MSCs also play a relevant role in the pathogenesis of hematological neoplasms. We aimed to identify overlapping mechanisms in MSCs derived from myeloid and lymphoid neoplasms contributing to disease progression and suppression of HSPCs to develop interventions that target these mechanisms. MSCs derived from healthy donors (n = 44) and patients diagnosed with myeloproliferative neoplasia (n = 11), myelodysplastic syndromes (n = 16), or acute myeloid leukemia (n = 25) and B-Non-Hodgkin lymphoma (n = 9) with BM infiltration and acute lymphoblastic leukemia (n = 9) were analyzed for their functionality and by RNA sequencing. A reduced growth and differentiation capacity of MSCs was found in all entities. RNA sequencing distinguished both groups but clearly showed overlapping differentially expressed genes, including major players in the BMP/TGF and WNT-signaling pathway which are crucial for growth, osteogenesis, and hematopoiesis. Functional alterations in healthy MSCs were inducible by exposure to supernatants from malignant cells, implicating the involvement of these factors in disease progression. Overall, we were able to identify overlapping factors that pose potential future therapeutic targets.

9.
Diagnostics (Basel) ; 13(5)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36899967

ABSTRACT

To date, only a single transcriptome-wide m6A sequencing study of clear cell renal cell carcinoma (ccRCC) has been reported, with no validation so far. Herein, by TCGA analysis of the KIRC cohort (n = 530 ccRCC; n = 72 normal), an external expression validation of 35 preidentified m6A targets was performed. Further in-depth expression stratification enabled assessment of m6A-driven key targets. Overall survival (OS) analysis and gene set enrichment analyses (GSEA) were conducted to assess their clinical and functional impact on ccRCC. In the hyper-up cluster significant upregulation was confirmed for NDUFA4L2, NXPH4, SAA1, and PLOD2 (40%) and in the hypo-up cluster for FCHSD1 (10%). Significant downregulation was observed for UMOD, ANK3, and CNTFR (27.3%) in the hypo-down cluster and for CHDH (25%) in the hyper-down cluster. In-depth expression stratification showed consistent dysregulation in ccRCC only for 11.67%: NDUFA4L2, NXPH4, and UMOD (NNU-panel). Patients with strong NNU panel dysregulation had significantly poorer OS (p = 0.0075). GSEA identified 13 associated and significantly upregulated gene sets (all p-values < 0.5; FDR < 0.25). External validation of the only available m6A sequencing in ccRCC consistently reduced dysregulated m6A-driven targets on the NNU panel with highly significant effects on OS. Epitranscriptomics are a promising target for developing novel therapies and for identifying prognostic markers for daily clinical practice.

10.
Mol Genet Genomics ; 287(3): 273-4, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22278756

ABSTRACT

MsLDR-creator is a free web service to design assays for the new DNA methylation detection method msLDR. The service provides the user with all necessary information about the oligonucleotides required for the measurement of a given CpG within a sequence of interest. The parameters are calculated by the nearest neighbour approach to achieve optimal behaviour during the experimental procedure. In addition, to guarantee a good start using msLDR, further information, like protocols and hints and tricks, are provided.


Subject(s)
CpG Islands , DNA Methylation , Sequence Analysis, DNA , Software , Computational Biology/methods , Internet
11.
Genome Med ; 14(1): 24, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35227293

ABSTRACT

BACKGROUND: Pancreatic neuroendocrine neoplasms (PanNENs) fall into two subclasses: the well-differentiated, low- to high-grade pancreatic neuroendocrine tumors (PanNETs), and the poorly-differentiated, high-grade pancreatic neuroendocrine carcinomas (PanNECs). While recent studies suggest an endocrine descent of PanNETs, the origin of PanNECs remains unknown. METHODS: We performed DNA methylation analysis for 57 PanNEN samples and found that distinct methylation profiles separated PanNENs into two major groups, clearly distinguishing high-grade PanNECs from other PanNETs including high-grade NETG3. DNA alterations and immunohistochemistry of cell-type markers PDX1, ARX, and SOX9 were utilized to further characterize PanNECs and their cell of origin in the pancreas. RESULTS: Phylo-epigenetic and cell-type signature features derived from alpha, beta, acinar, and ductal adult cells suggest an exocrine cell of origin for PanNECs, thus separating them in cell lineage from other PanNENs of endocrine origin. CONCLUSIONS: Our study provides a robust and clinically applicable method to clearly distinguish PanNECs from G3 PanNETs, improving patient stratification.


Subject(s)
Carcinoma, Neuroendocrine , Neuroendocrine Tumors , Pancreatic Neoplasms , Adult , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , DNA Methylation , Humans , Neoplasm Grading , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology
12.
Mol Genet Genomics ; 286(3-4): 279-91, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21879293

ABSTRACT

A new sensitive method for multiplex gene-specific methylation analysis was developed using a ligation-based approach combined with a TaqMan-based detection and readout employing universal reporter probes. The approach, termed methylation-specific Ligation Detection Reaction (msLDR), was applied to test 16 loci in 8 different colorectal cancer cells in parallel. These loci encode immune regulatory genes involved in T-cell and natural killer cell activation, whose silencing is associated with the development or progression of colorectal cancer. Parallel analysis of HLA-A, HLA-B, STAT1, B2M, LMP2, LMP7, PA28α, TAP1, TAP2, TAPBP, ULBP2 and ULBP3 by msLDR in eight colorectal cancer cell lines showed preferential methylation at the HLA-B, ULBP2 and ULBB3 loci, but not at the other loci. MsLDR was found to represent a suitable and sensitive method for the detection of distinct methylation patterns as validated by conventional bisulphite Sanger sequencing and COBRA analysis. Since gene silencing by epigenetic mechanisms plays a central role during transformation of a normal differentiated somatic cell into a cancer cell, characterization of the gene methylation status in tumours is a major topic not only in basic research, but also in clinical diagnostics. Due to a very simple workflow, msLDR is likely to be applicable to clinical samples and thus comprises a potential diagnostic tool for clinical purposes.


Subject(s)
DNA Methylation , Genetic Techniques , Antigen Presentation/genetics , Cell Line, Tumor , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , GPI-Linked Proteins/genetics , Genes, MHC Class I , Genetic Techniques/statistics & numerical data , Humans , Intercellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , Miniaturization , Polymerase Chain Reaction/methods , T-Lymphocytes/immunology
13.
Epigenetics ; 15(12): 1319-1324, 2020 12.
Article in English | MEDLINE | ID: mdl-32475296

ABSTRACT

Merkel cell carcinoma (MCC) is a very rare, but highly aggressive skin cancer which occurs mainly in elderly patients. MCC cells show an expression pattern of three cell lineages: epithelial, neuroendocrine, and B-cell progenitor. This trilinear expression pattern suggests stemness activity in MCC. The etiopathogenesis of MCC is either linked to the Merkel cell polyomavirus (MCPyV) or in a smaller proportion (20%) to high levels of UV-induced somatic mutations. Both viral presence and accumulation of mutations have been shown to be associated with accelerated DNA methylation Age (DNAmAge) compared to chronological age. The MCC DNAmAge was significantly lower compared to the chronological age, which was irrespective of the viral presence or mutational burden. Although these features indicate some aspects of stemness in MCC cells, gene-expression-based pluripotency testing did not provide evidence for pluripotency of MCC cells.


Subject(s)
Carcinoma, Merkel Cell/genetics , Cellular Senescence , Epigenesis, Genetic , Mutation Accumulation , Aged , Aged, 80 and over , Carcinoma, Merkel Cell/pathology , Carcinoma, Merkel Cell/virology , DNA Methylation , Female , Humans , Male , Merkel cell polyomavirus/pathogenicity , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology
14.
Genome Med ; 12(1): 46, 2020 05 25.
Article in English | MEDLINE | ID: mdl-32450911

ABSTRACT

BACKGROUND: Hutchinson-Gilford progeria syndrome (HGPS) is a progeroid disease characterized by the early onset of age-related phenotypes including arthritis, loss of body fat and hair, and atherosclerosis. Cells from affected individuals express a mutant version of the nuclear envelope protein lamin A (termed progerin) and have previously been shown to exhibit prominent histone modification changes. METHODS: Here, we analyze the possibility that epigenetic deregulation of lamina-associated domains (LADs) is involved in the molecular pathology of HGPS. To do so, we studied chromatin accessibility (Assay for Transposase-accessible Chromatin (ATAC)-see/-seq), DNA methylation profiles (Infinium MethylationEPIC BeadChips), and transcriptomes (RNA-seq) of nine primary HGPS fibroblast cell lines and six additional controls, two parental and four age-matched healthy fibroblast cell lines. RESULTS: Our ATAC-see/-seq data demonstrate that primary dermal fibroblasts from HGPS patients exhibit chromatin accessibility changes that are enriched in LADs. Infinium MethylationEPIC BeadChip profiling further reveals that DNA methylation alterations observed in HGPS fibroblasts are similarly enriched in LADs and different from those occurring during healthy aging and Werner syndrome (WS), another premature aging disease. Moreover, HGPS patients can be stratified into two different subgroups according to their DNA methylation profiles. Finally, we show that the epigenetic deregulation of LADs is associated with HGPS-specific gene expression changes. CONCLUSIONS: Taken together, our results strongly implicate epigenetic deregulation of LADs as an important and previously unrecognized feature of HGPS, which contributes to disease-specific gene expression. Therefore, they not only add a new layer to the study of epigenetic changes in the progeroid syndrome, but also advance our understanding of the disease's pathology at the cellular level.


Subject(s)
Lamin Type A/genetics , Progeria/genetics , Cell Line , DNA Methylation , Epigenesis, Genetic , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Protein Domains
15.
Nat Commun ; 11(1): 4974, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33009381

ABSTRACT

Generation of bispecific antibodies (bsAbs) requires a combination of compatible binders in formats that support desired functionalities. Here, we report that bsAb-matrices can be generated by Format Chain Exchange (FORCE), enabling screening of combinatorial binder/format spaces. Input molecules for generation of bi/multi-valent bsAbs are monospecific entities similar to knob-into-hole half-antibodies, yet with complementary CH3-interface-modulated and affinity-tagged dummy-chains. These contain mutations that lead to limited interface repulsions without compromising expression or biophysical properties of educts. Mild reduction of combinations of educts triggers spontaneous chain-exchange reactions driven by partially flawed CH3-educt interfaces resolving to perfect complementarity. This generates large bsAb matrices harboring different binders in multiple formats. Benign biophysical properties and good expression yields of educts, combined with simplicity of purification enables process automation. Examples that demonstrate the relevance of screening binder/format combinations are provided as a matrix of bsAbs that simultaneously bind Her1/Her2 and DR5 without encountering binder or format-inflicted interferences.


Subject(s)
Antibodies, Bispecific/biosynthesis , High-Throughput Screening Assays , Antibodies, Bispecific/isolation & purification , Automation , HEK293 Cells , Humans , Mutation/genetics , Protein Multimerization
16.
Cell Rep ; 23(11): 3407-3418, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29898408

ABSTRACT

Colorectal adenomas are precursor lesions of colorectal cancers and represent clonal amplifications of single cells from colonic crypts. DNA methylation patterns specify cell-type identity during cellular differentiation and, therefore, provide opportunities for the molecular analysis of tumors. We have now analyzed DNA methylation patterns in colorectal adenomas and identified three biologically defined subclasses that describe different intestinal crypt differentiation stages. Importantly, colorectal carcinomas could be classified into the same methylation subtypes, reflecting their shared cell types of origin with adenomas. Further data analysis also revealed significantly reduced overall survival for one of the subtypes. Our results provide a concept for understanding the methylation patterns observed in colorectal cancer and provide opportunities for tumor subclassification and patient stratification.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Adenoma/classification , Adenoma/genetics , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/classification , Colorectal Neoplasms/genetics , Epigenomics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Transcription Factors/metabolism
17.
Nat Commun ; 9(1): 577, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422656

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer and usually progresses from a UV-induced precancerous lesion termed actinic keratosis (AK). Despite various efforts to characterize these lesions molecularly, the etiology of AK and its progression to cSCC remain partially understood. Here, we use Infinium MethylationEPIC BeadChips to interrogate the DNA methylation status in healthy, AK and cSCC epidermis samples. Importantly, we show that AK methylation patterns already display classical features of cancer methylomes and are highly similar to cSCC profiles. Further analysis identifies typical features of stem cell methylomes, such as reduced DNA methylation age, non-CpG methylation, and stem cell-related keratin and enhancer methylation patterns. Interestingly, this signature is detected only in half of the samples, while the other half shows patterns more closely related to healthy epidermis. These findings suggest the existence of two subclasses of AK and cSCC emerging from distinct keratinocyte differentiation stages.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Keratosis, Actinic/genetics , Skin Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Differentiation , Female , Humans , Keratinocytes , Male , Middle Aged , Young Adult
18.
Cold Spring Harb Mol Case Stud ; 3(3): a001610, 2017 05.
Article in English | MEDLINE | ID: mdl-28487883

ABSTRACT

Colon polyps represent precursor lesions of colon cancers and their malignant potential varies according to histological subtype. A rare subtype of colon polyps is the Peutz-Jeghers (PJ) polyp. PJ polyps mostly occur in the context of Peutz-Jeghers syndrome, which is characterized by the development of multiple polyps in the intestinal tract and hyperpigmentation of oral mucosa and lips. Peutz-Jeghers is an autosomal dominant disorder caused by pathogenic variants of the serine threonine kinase STK11. PJ polyps very rarely occur outside of the syndrome and are then referred to as solitary PJ polyps. Contrary to the situation in Peutz-Jeghers, the genetic basis and the malignant potential of solitary PJ polyps are currently unknown. Here we describe a detailed and comprehensive genetic profile of a solitary PJ polyp. Pathological examination revealed a high tissue homogeneity with >80% epithelial cells. Whole-genome sequencing failed to identify any clonal mutations but demonstrated a significant number of subclonal mutations. No somatic or germline mutations were found at the STK11 locus, suggesting that solitary PJ polyps are genetically distinct from Peutz-Jeghers polyps. In addition, methylome analysis revealed global hypomethylation and CpG island hypermethylation, two features that have been described as hallmarks of the colorectal cancer epigenome. These results provide an example of a premalignant lesion that is defined by epigenetic, rather than genetic changes. Furthermore, our findings support the notion that solitary PJ polyps constitute neoplastic tissue with malignant potential that should be removed for cancer prevention.


Subject(s)
Colonic Polyps/genetics , Hamartoma/genetics , Peutz-Jeghers Syndrome/genetics , Adult , Colon/pathology , Colonic Polyps/pathology , Germ-Line Mutation , Humans , Male , Mutation , Peutz-Jeghers Syndrome/diagnosis , Whole Genome Sequencing
19.
Aging Cell ; 15(3): 563-71, 2016 06.
Article in English | MEDLINE | ID: mdl-27004597

ABSTRACT

Epigenetic changes represent an attractive mechanism for understanding the phenotypic changes associated with human aging. Age-related changes in DNA methylation at the genome scale have been termed 'epigenetic drift', but the defining features of this phenomenon remain to be established. Human epidermis represents an excellent model for understanding age-related epigenetic changes because of its substantial cell-type homogeneity and its well-known age-related phenotype. We have now generated and analyzed the currently largest set of human epidermis methylomes (N = 108) using array-based profiling of 450 000 methylation marks in various age groups. Data analysis confirmed that age-related methylation differences are locally restricted and characterized by relatively small effect sizes. Nevertheless, methylation data could be used to predict the chronological age of sample donors with high accuracy. We also identified discontinuous methylation changes as a novel feature of the aging methylome. Finally, our analysis uncovered an age-related erosion of DNA methylation patterns that is characterized by a reduced dynamic range and increased heterogeneity of global methylation patterns. These changes in methylation variability were accompanied by a reduced connectivity of transcriptional networks. Our findings thus define the loss of epigenetic regulatory fidelity as a key feature of the aging epigenome.


Subject(s)
DNA Methylation/genetics , Gene Regulatory Networks/genetics , Skin Aging/genetics , Transcription, Genetic , Adolescent , Adult , Aged , Epigenesis, Genetic , Humans , Middle Aged , Models, Biological , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL