Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Methods Mol Biol ; 2621: 91-109, 2023.
Article in English | MEDLINE | ID: mdl-37041442

ABSTRACT

Microsatellites are short tandem repeats of one to six nucleotides that are highly polymorphic and extensively used as genetic markers in numerous biomedical applications, including the detection of microsatellite instability (MSI) in cancer. The standard analytical method for microsatellite analysis relies on PCR amplification followed by capillary electrophoresis or, more recently, next-generation sequencing (NGS). However, their amplification during PCR generates undesirable frameshift products known as stutter peaks caused by polymerase slippage, complicating data analysis and interpretation, while very few alternative methods for microsatellite amplification have been developed to reduce the formation of these artifacts. In this context, the recently developed low-temperature recombinase polymerase amplification (LT-RPA) is an isothermal DNA amplification method at low temperature (32 °C) that drastically reduces and sometimes completely abolishes the formation of stutter peaks. LT-RPA greatly simplifies the genotyping of microsatellites and improves the detection of MSI in cancer. In this chapter, we describe in detail all the experimental steps necessary for the development of LT-RPA simplex and multiplex assays for microsatellite genotyping and MSI detection, including the design, optimization, and validation of the assays combined with capillary electrophoresis or NGS.


Subject(s)
Microsatellite Instability , Neoplasms , Humans , Recombinases/genetics , Genotype , Microsatellite Repeats , DNA/genetics , Nucleotidyltransferases , Neoplasms/genetics
2.
Aging (Albany NY) ; 14(19): 7718-7733, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36202132

ABSTRACT

Aging is a progressive time-dependent biological process affecting differentially individuals, who can sometimes present exceptional longevity. Epigenetic alterations are one of the hallmarks of aging, which comprise the epigenetic drift and clock at DNA methylation level. In the present study, we estimated the DNA methylation-based age (DNAmage) using four epigenetic clocks based on a small number of CpGs in French centenarians and semi-supercentenarians (CSSC, n=214) as well as nonagenarians' and centenarians' offspring (NCO, n=143) compared to individuals from the French general population (CG, n=149). DNA methylation analysis of the nine CpGs included in the epigenetic clocks showed high correlation with chronological age (-0.66>R>0.54) and also the presence of an epigenetic drift for four CpGs that was only visible in CSSC. DNAmage analysis showed that CSSC and to a lesser extend NCO present a younger DNAmage than their chronological age (15-28.5 years for CSSC, 4.4-11.5 years for NCO and 4.2-8.2 years for CG), which were strongly significant in CSSC compared to CG (p-values<2.2e-16). These differences suggest that epigenetic aging and potentially biological aging are slowed in exceptionally long-lived individuals and that epigenetic clocks based on a small number of CpGs are sufficient to reveal alterations of the global epigenetic clock.


Subject(s)
Centenarians , Epigenesis, Genetic , Aged, 80 and over , Humans , CpG Islands/genetics , Epigenomics , DNA Methylation , Aging/genetics
3.
Elife ; 112022 09 20.
Article in English | MEDLINE | ID: mdl-36125262

ABSTRACT

Aberrant DNA methylation is a well-known feature of tumours and has been associated with metastatic melanoma. However, since melanoma cells are highly heterogeneous, it has been challenging to use affected genes to predict tumour aggressiveness, metastatic evolution, and patients' outcomes. We hypothesized that common aggressive hypermethylation signatures should emerge early in tumorigenesis and should be shared in aggressive cells, independent of the physiological context under which this trait arises. We compared paired melanoma cell lines with the following properties: (i) each pair comprises one aggressive counterpart and its parental cell line and (ii) the aggressive cell lines were each obtained from different host and their environment (human, rat, and mouse), though starting from the same parent cell line. Next, we developed a multi-step genomic pipeline that combines the DNA methylome profile with a chromosome cluster-oriented analysis. A total of 229 differentially hypermethylated genes was commonly found in the aggressive cell lines. Genome localization analysis revealed hypermethylation peaks and clusters, identifying eight hypermethylated gene promoters for validation in tissues from melanoma patients. Five Cytosine-phosphate-Guanine (CpGs) identified in primary melanoma tissues were transformed into a DNA methylation score that can predict survival (log-rank test, p=0.0008). This strategy is potentially universally applicable to other diseases involving DNA methylation alterations.


Subject(s)
Melanoma , Skin Neoplasms , Animals , Chromosomes , CpG Islands , Cytosine , DNA Methylation , Epigenesis, Genetic , Epigenome , Gene Expression Regulation, Neoplastic , Guanine , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Phosphates , Rats , Skin Neoplasms/genetics , Melanoma, Cutaneous Malignant
4.
Sci Rep ; 12(1): 4684, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35304543

ABSTRACT

Lymphoblastoid cell lines (LCLs) derive from blood infected in vitro by Epstein-Barr virus and were used in several genetic, transcriptomic and epigenomic studies. Although few changes were shown between LCL and blood genotypes (SNPs) validating their use in genetics, more were highlighted for other genomic features and/or in their transcriptome and epigenome. This could render them less appropriate for these studies, notably when blood DNA could still be available. Here we developed a simple, high-throughput and cost-effective real-time PCR approach allowing to distinguish blood from LCL DNA samples based on the presence of EBV relative load and rearranged T-cell receptors γ and ß. Our approach was able to achieve 98.5% sensitivity and 100% specificity on DNA of known origin (458 blood and 316 LCL DNA). It was further applied to 1957 DNA samples from the CEPH Aging cohort comprising DNA of uncertain origin, identifying 784 blood and 1016 LCL DNA. A subset of these DNA was further analyzed with an epigenetic clock indicating that DNA extracted from blood should be preferred to LCL for DNA methylation-based age prediction analysis. Our approach could thereby be a powerful tool to ascertain the origin of DNA in old collections prior to (epi)genomic studies.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Cell Line , DNA/genetics , Epigenomics , Herpesvirus 4, Human/genetics , Humans , Real-Time Polymerase Chain Reaction
5.
Sci Rep ; 10(1): 15652, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32973211

ABSTRACT

Several blood-based age prediction models have been developed using less than a dozen to more than a hundred DNA methylation biomarkers. Only one model (Z-P1) based on pyrosequencing has been developed using DNA methylation of a single locus located in the ELOVL2 promoter, which is considered as one of the best age-prediction biomarker. Although multi-locus models generally present better performances compared to the single-locus model, they require more DNA and present more inter-laboratory variations impacting the predictions. Here we developed 17,018 single-locus age prediction models based on DNA methylation of the ELOVL2 promoter from pooled data of four different studies (training set of 1,028 individuals aged from 0 and 91 years) using six different statistical approaches and testing every combination of the 7 CpGs, aiming to improve the prediction performances and reduce the effects of inter-laboratory variations. Compared to Z-P1 model, three statistical models with the optimal combinations of CpGs presented improved performances (MAD of 4.41-4.77 in the testing set of 385 individuals) and no age-dependent bias. In an independent testing set of 100 individuals (19-65 years), we showed that the prediction accuracy could be further improved by using different CpG combinations and increasing the number of technical replicates (MAD of 4.17).


Subject(s)
Aging/blood , Aging/genetics , DNA Methylation , Fatty Acid Elongases/genetics , Genetic Loci/genetics , Laboratories , Promoter Regions, Genetic/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , CpG Islands/genetics , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Young Adult
6.
Oncol Lett ; 20(2): 1999-2006, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32724446

ABSTRACT

DNA hypomethylation of long interspersed repetitive DNA retrotransposon (LINE-1) and Alu repeats elements of short interspersed elements family (SINEs) is an early event in carcinogenesis that causes transcriptional activation and leads to chromosomal instability. In the current study, DNA methylation levels of LINE-1 and Alu repeats were analyzed in tumoral tissues of invasive breast cancer in a Tunisian cohort and its association with the clinicopathological features of patients was defined. DNA methylation of LINE-1 and Alu repeats were analyzed using pyrosequencing in 61 invasive breast cancers. Median values observed for DNA methylation of LINE-1 and Alu repeats were considered as the cut-off (59.81 and 18.49%, respectively). The results of the current study demonstrated a positive correlation between DNA methylation levels of LINE-1 and Alu repeats (rho=0.284; P<0.03). DNA hypomethylation of LINE-1 was also indicated to be associated with low grade (P=0.023). To the best of our knowledge, the current study is the first study regarding DNA methylation of LINE-1 and Alu repeats element in breast cancer of the Tunisian population. The results of the current study suggest that, since hypomethylation of LINE-1 is associated with low grade, it could be used as a biomarker for prognosis for patients with breast cancer.

7.
Nucleic Acids Res ; 47(21): e141, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31584085

ABSTRACT

Microsatellites are polymorphic short tandem repeats of 1-6 nucleotides ubiquitously present in the genome that are extensively used in living organisms as genetic markers and in oncology to detect microsatellite instability (MSI). While the standard analysis method of microsatellites is based on PCR followed by capillary electrophoresis, it generates undesirable frameshift products known as 'stutter peaks' caused by the polymerase slippage that can greatly complicate the analysis and interpretation of the data. Here we present an easy multiplexable approach replacing PCR that is based on low temperature isothermal amplification using recombinase polymerase amplification (LT-RPA) that drastically reduces and sometimes completely abolishes the formation of stutter artifacts, thus greatly simplifying the calling of the alleles. Using HT17, a mononucleotide DNA repeat that was previously proposed as an optimal marker to detect MSI in tumor DNA, we showed that LT-RPA improves the limit of detection of MSI compared to PCR up to four times, notably for small deletions, and simplifies the identification of the mutant alleles. It was successfully applied to clinical colorectal cancer samples and enabled detection of MSI. This easy-to-handle, rapid and cost-effective approach may deeply improve the analysis of microsatellites in several biological and clinical applications.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms , DNA/genetics , Microsatellite Instability , Microsatellite Repeats/genetics , Nucleic Acid Amplification Techniques/methods , Cell Line, Tumor , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Humans , Temperature
8.
Sci Rep ; 9(1): 8862, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31222117

ABSTRACT

DNA methylation has been identified as the most promising molecular biomarker for the prediction of age. Several DNA methylation-based models have been proposed for age prediction based on blood samples, using mainly pyrosequencing. These methods present different performances for age prediction and have rarely, if ever, been evaluated and intercompared in an independent validation study. Here, for the first time, we evaluate and compare six blood-based age prediction models (Bekaert1, Park2, Thong3, Weidner4, and the Zbiec-Piekarska 15 and Zbiec-Piekarska 26), using DNA methylation analysis by pyrosequencing on 100 blood samples from French individuals aged between 19-65 years. For each model, we perform correlation analysis and evaluate age-prediction performance (mean absolute deviation (MAD) and standard error of the estimate (SEE)). The best age-prediction performances were found with the Bekaert and Thong models (MAD of 4.5-5.2, SEE of 6.8-7.2), followed by the Zbiec-Piekarska 1 model (MAD of 6.8 and SEE of 9.2), while the Park, Weidner and Zbiec-Piekarska 2 models presented lower performances (MAD of 7.2-8.7 and SEE of 9.2-10.3). Given these results, we recommend performing systematic, independent evaluation of all age prediction models on a same cohort to validate the different models and compare their performance.


Subject(s)
Aging/genetics , DNA Methylation , DNA/blood , Adult , Aged , Female , Genetic Markers , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Models, Statistical , Young Adult
9.
J Invest Dermatol ; 139(9): 2004-2015.e13, 2019 09.
Article in English | MEDLINE | ID: mdl-31059696

ABSTRACT

The management of large congenital melanocytic nevi (lCMN) is based exclusively on iterative surgical procedures in the absence of validated medical therapy. The aim of our study was to develop an intra-lesional medical treatment for lCMN. Seventeen patients harboring NRAS-mutated lCMN were included. Nevocytes obtained from lCMN displayed an overactivation of mitogen-activated protein kinase and phosphoinositide 3-kinase (Akt) pathways. Mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) and Akt inhibitors reduced the nevosphere diameter in sphere-forming assays, as well as cell viability and proliferation in in vitro assays. Standardized lCMN explants were then cultured ex vivo with the same inhibitors, which induced a decrease in MelanA+ and Sox10+ cells in both epidermis and dermis. Finally, intradermal injections of these inhibitors were administered within standardized lCMN xenografts in Rag2-/- mice. They induced a dramatic decrease in nevocytes in treated xenografts, which persisted 30 days after the end of treatment. Using original nevus explant and xenograft preclinical models, we demonstrated that intradermal MEK/Akt inhibition might serve as neoadjuvant therapy for the treatment of NRAS-mutated congenital melanocytic nevi to avoid iterative surgeries.


Subject(s)
Antineoplastic Agents/administration & dosage , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nevus, Pigmented/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Proliferation/drug effects , Child , Child, Preschool , Female , GTP Phosphohydrolases/genetics , Humans , Infant , Injections, Intradermal , Injections, Intralesional , MART-1 Antigen/metabolism , Male , Melanocytes/drug effects , Melanocytes/pathology , Membrane Proteins/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Nevus, Pigmented/congenital , Nevus, Pigmented/genetics , Nevus, Pigmented/pathology , Proto-Oncogene Proteins c-akt/metabolism , SOXE Transcription Factors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Skin/cytology , Skin/pathology , Skin Neoplasms/congenital , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Proc Natl Acad Sci U S A ; 116(13): 6298-6307, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30846549

ABSTRACT

Natural regulatory T cells (nTregs) ensure the control of self-tolerance and are currently used in clinical trials to alleviate autoimmune diseases and graft-versus-host disease after hematopoietic stem cell transfer. Based on CD39/CD26 markers, blood nTreg analysis revealed the presence of five different cell subsets, each representing a distinct stage of maturation. Ex vivo added microenvironmental factors, including IL-2, TGFß, and PGE2, direct the conversion from naive precursor to immature memory and finally from immature to mature memory cells, the latest being a no-return stage. Phenotypic and genetic characteristics of the subsets illustrate the structural parental maturation between subsets, which further correlates with the expression of regulatory factors. Regarding nTreg functional plasticity, both maturation stage and microenvironmental cytokines condition nTreg activities, which include blockade of autoreactive immune cells by cell-cell contact, Th17 and IL-10 Tr1-like activities, or activation of TCR-stimulating dendritic cell tolerization. Importantly, blood nTreg CD39/CD26 profile remained constant over a 2-y period in healthy persons but varied from person to person. Preliminary data on patients with autoimmune diseases or acute myelogenous leukemia illustrate the potential use of the nTreg CD39/CD26 profile as a blood biomarker to monitor chronic inflammatory diseases. Finally, we confirmed that naive conventional CD4 T cells, TCR-stimulated under a tolerogenic conditioned medium, could be ex vivo reprogrammed to FOXP3 lineage Tregs, and further found that these cells were exclusively committed to suppressive function under all microenvironmental contexts.


Subject(s)
Cellular Microenvironment/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Apyrase/blood , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dinoprostone/metabolism , Dipeptidyl Peptidase 4/blood , Forkhead Transcription Factors/metabolism , Humans , Interleukin-10/metabolism , Interleukin-2/metabolism , Leukemia, Myeloid , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism
11.
Clin Epigenetics ; 11(1): 9, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30651148

ABSTRACT

BACKGROUND: Efficient treatments against metastatic melanoma dissemination are still lacking. Here, we report that low-cytotoxic concentrations of 5-aza-2'-deoxycytidine, a DNA demethylating agent, prevent in vitro 3D invasiveness of metastatic melanoma cells and reduce lung metastasis formation in vivo. RESULTS: We unravelled that this beneficial effect is in part due to MIR-199A2 re-expression by promoter demethylation. Alone, this miR showed an anti-invasive and anti-metastatic effect. Throughout integration of micro-RNA target prediction databases with transcriptomic analysis after 5-aza-2'-deoxycytidine treatments, we found that miR-199a-3p downregulates set of genes significantly involved in invasion/migration processes. In addition, analysis of data from melanoma patients showed a stage- and tissue type-dependent modulation of MIR-199A2 expression by DNA methylation. CONCLUSIONS: Thus, our data suggest that epigenetic- and/or miR-based therapeutic strategies can be relevant to limit metastatic dissemination of melanoma.


Subject(s)
DNA Methylation/drug effects , Decitabine/pharmacology , Lung Neoplasms/secondary , Melanoma/genetics , MicroRNAs/genetics , Spheroids, Cellular/cytology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Melanoma/drug therapy , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Promoter Regions, Genetic , Sequence Analysis, RNA , Spheroids, Cellular/drug effects , Up-Regulation
13.
Hum Mutat ; 39(3): 441-453, 2018 03.
Article in English | MEDLINE | ID: mdl-29227006

ABSTRACT

Every colorectal cancer (CRC) patient should be tested for microsatellite instability (MSI) to screen for Lynch syndrome. Evaluation of MSI status involves screening tumor DNA for the presence of somatic deletions in DNA repeats using PCR followed by fragment analysis. While this method may lack sensitivity due to the presence of a high level of germline DNA, which frequently contaminates the core of primary colon tumors, no other method developed to date is capable of modifying the standard PCR protocol to achieve improvement of MSI detection. Here, we describe a new approach developed for the ultra-sensitive detection of MSI in CRC based on E-ice-COLD-PCR, using HSP110 T17, a mononucleotide DNA repeat previously proposed as an optimal marker to detect MSI in tumor DNA, and an oligo(dT)16 LNA blocker probe complementary to wild-type genotypes. The HT17 E-ice-COLD-PCR assay improved MSI detection by 20-200-fold compared with standard PCR using HT17 alone. It presents an analytical sensitivity of 0.1%-0.05% of mutant alleles in wild-type background, thus greatly improving MSI detection in CRC samples highly contaminated with normal DNA. HT17 E-ice-COLD-PCR is a rapid, cost-effective, easy-to-implement, and highly sensitive method, which could significantly improve the detection of MSI in routine clinical testing.


Subject(s)
Colorectal Neoplasms/genetics , HSP110 Heat-Shock Proteins/genetics , Microsatellite Instability , Polymerase Chain Reaction/methods , Cell Line, Tumor , Cold Temperature , Germ Cells/metabolism , Humans , Mutation/genetics , Reference Standards
14.
Oncotarget ; 7(39): 63690-63707, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27577073

ABSTRACT

HLA-G is an immune checkpoint molecule with specific relevance in cancer immunotherapy. It was first identified in cytotrophoblasts, protecting the fetus from maternal rejection. HLA-G tissue expression is very restricted but induced in numerous malignant tumors such as glioblastoma, contributing to their immune escape. Hypoxia occurs during placenta and tumor development and was shown to activate HLA-G. We aimed to elucidate the mechanisms of HLA-G activation under conditions combining hypoxia-mimicking treatment and 5-aza-2'deoxycytidine, a DNA demethylating agent used in anti-cancer therapy which also induces HLA-G. Both treatments enhanced the amount of HLA-G mRNA and protein in HLA-G negative U251MG glioma cells. Electrophoretic Mobility Shift Assays and luciferase reporter gene assays revealed that HLA-G upregulation depends on Hypoxia Inducible Factor-1 (HIF-1) and a hypoxia responsive element (HRE) located in exon 2. A polymorphic HRE at -966 bp in the 5'UT region may modulate the magnitude of the response mediated by the exon 2 HRE. We suggest that therapeutic strategies should take into account that HLA-G expression in response to hypoxic tumor environment is dependent on HLA-G gene polymorphism and DNA methylation state at the HLA-G locus.


Subject(s)
Glioma/immunology , HLA-G Antigens/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Response Elements , 5' Untranslated Regions , Antineoplastic Agents/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , CpG Islands , DNA Methylation , Decitabine , Exons , Genes, MHC Class I , Humans , Hypoxia , Immune System , Neoplasm Invasiveness , Polymorphism, Genetic , Promoter Regions, Genetic , Transcriptional Activation
16.
Mol Oncol ; 10(2): 330-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26601720

ABSTRACT

BACKGROUND: DNA methylation alterations are early events in tumorigenesis and important in the regulation of gene expression in cancer cells. Lung cancer patients have in general a poor prognosis, and a deeper insight into the epigenetic landscape in lung adenocarcinoma tumors and its prognostic implications is needed. RESULTS: We determined whole-genome DNA methylation profiles of 164 fresh frozen lung adenocarcinoma samples and 19 samples of matched normal lung tissue using the Illumina Infinium 450K array. A large number of differentially methylated CpGs in lung adenocarcinoma tissue were identified, and specific methylation profiles were observed in tumors with mutations in the EGFR-, KRAS- or TP53 genes and according to the patients' smoking status. The methylation levels were correlated with gene expression and both positive and negative correlations were seen. Methylation profiles of the tumor samples identified subtypes of tumors with distinct prognosis, including one subtype enriched for TP53 mutant tumors. A prognostic index based on the methylation levels of 33 CpGs was established, and was significantly associated with prognosis in the univariate analysis using an independent cohort of lung adenocarcinoma patients from The Cancer Genome Atlas project. CpGs in the HOX B and HOX C gene clusters were represented in the prognostic signature. CONCLUSIONS: Methylation differences mirror biologically important features in the etiology of lung adenocarcinomas and influence prognosis.


Subject(s)
Adenocarcinoma/genetics , DNA Methylation , ErbB Receptors/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Cohort Studies , CpG Islands , Female , Gene Expression Regulation, Neoplastic , Genes, Homeobox , Genome, Human , Humans , Male , Middle Aged , Mutation , Norway/epidemiology , Prognosis , Signal Transduction , Smoking/adverse effects , Smoking/genetics , TOR Serine-Threonine Kinases/metabolism
17.
Endocr Relat Cancer ; 22(6): 953-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26400872

ABSTRACT

Overexpression of insulin growth factor 2 (IGF2) is a hallmark of adrenocortical carcinomas and pheochromocytomas. Previous studies investigating the IGF2/H19 locus have mainly focused on a single molecular level such as genomic alterations or altered DNA methylation levels and the causal changes underlying IGF2 overexpression are still not fully established. In the current study, we analyzed 62 tumors of the adrenal gland from patients with Conn's adenoma (CA, n=12), pheochromocytomas (PCC, n=10), adrenocortical benign tumors (ACBT, n=20), and adrenocortical carcinomas (ACC, n=20). Gene expression, somatic copy number variation of chr11p15.5, and DNA methylation status of three differential methylated regions of the IGF2/H19 locus including the H19 imprinting control region were integratively analyzed. IGF2 overexpression was found in 85% of the ACCs and 100% of the PCCs compared to 23% observed in CAs and ACBTs. Copy number aberrations of chr11p15.5 were abundant in both PCCs and ACCs but while PCCs retained a diploid state, ACCs were frequently tetraploid (7/19). Loss of either a single allele or loss of two alleles of the same parental origin in tetraploid samples resulted in a uniparental disomy-like genotype. These copy number changes correlated with hypermethylation of the H19 ICR suggesting that the lost alleles were the unmethylated maternal alleles. Our data provide conclusive evidence that loss of the maternal allele correlates with IGF2 overexpression in adrenal tumors and that hypermethylation of the H19 ICR is a consequence thereof.


Subject(s)
Adrenal Gland Neoplasms/genetics , Adrenocortical Adenoma/genetics , Carcinoma/genetics , DNA Methylation , Gene Dosage , Gene Expression Regulation, Neoplastic/genetics , Insulin-Like Growth Factor II/genetics , Neoplasm Proteins/genetics , Pheochromocytoma/genetics , Adrenal Gland Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Adult , Aged , Alleles , Carcinoma/metabolism , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/ultrastructure , Female , Genomic Imprinting , Genotype , Humans , Insulin-Like Growth Factor II/biosynthesis , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Pheochromocytoma/metabolism , Ploidies , Polymorphism, Single Nucleotide , Up-Regulation
18.
Methods Mol Biol ; 1315: 315-33, 2015.
Article in English | MEDLINE | ID: mdl-26103908

ABSTRACT

Interaction and co-occurrence of protein and DNA-based epigenetic modifications have become a topic of interest for many fundamental and biomedical questions. We describe within this chapter a protocol that combines two techniques in order to determine the methylation status of the DNA specifically associated with a protein of interest. First, DNA that directly interacts with the selected protein (such as a specific histone modification, a transcription factor, or any other DNA-associated protein) is purified by standard chromatin immunoprecipitation (ChIP). Second, the level of DNA methylation of this immunoprecipitated DNA is measured by bisulfite conversion and Pyrosequencing, a quantitative sequencing-by-synthesis method. This procedure allows determining the methylation status of genomic DNA associated to a specific protein at single nucleotide resolution.


Subject(s)
Chromatin Immunoprecipitation/methods , DNA Methylation , Nucleotides/genetics , Sequence Analysis, DNA/methods , Analytic Sample Preparation Methods , Cell Line, Tumor , DNA/genetics , DNA/isolation & purification , DNA Methylation/drug effects , DNA Primers/genetics , Humans , Microspheres , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Sulfites/pharmacology
19.
Plant Mol Biol ; 88(4-5): 471-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26072424

ABSTRACT

Pyrosequencing permits accurate quantification of DNA methylation of specific regions where the proportions of the C/T polymorphism induced by sodium bisulfite treatment of DNA reflects the DNA methylation level. The commercially available high-throughput locus-specific pyrosequencing instruments allow for the simultaneous analysis of 96 samples, but restrict the DNA methylation analysis to CpG dinucleotide sites, which can be limiting in many biological systems. In contrast to mammals where DNA methylation occurs nearly exclusively on CpG dinucleotides, plants genomes harbor DNA methylation also in other sequence contexts including CHG and CHH motives, which cannot be evaluated by these pyrosequencing instruments due to software limitations. Here, we present a complete pipeline for accurate CpG and non-CpG cytosine methylation analysis at single base-resolution using high-throughput locus-specific pyrosequencing. The devised approach includes the design and validation of PCR amplification on bisulfite-treated DNA and pyrosequencing assays as well as the quantification of the methylation level at every cytosine from the raw peak intensities of the Pyrograms by two newly developed Visual Basic Applications. Our method presents accurate and reproducible results as exemplified by the cytosine methylation analysis of the promoter regions of two Tomato genes (NOR and CNR) encoding transcription regulators of fruit ripening during different stages of fruit development. Our results confirmed a significant and temporally coordinated loss of DNA methylation on specific cytosines during the early stages of fruit development in both promoters as previously shown by WGBS. The manuscript describes thus the first high-throughput locus-specific DNA methylation analysis in plants using pyrosequencing.


Subject(s)
DNA Methylation , DNA, Plant/genetics , DNA, Plant/metabolism , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Solanum lycopersicum/genetics , Solanum lycopersicum/metabolism , Base Sequence , CpG Islands , Cytosine/metabolism , DNA Primers/genetics , DNA, Chloroplast/genetics , Genes, Plant , High-Throughput Nucleotide Sequencing/statistics & numerical data , Promoter Regions, Genetic , Sequence Analysis, DNA/statistics & numerical data , Software , Sulfites
20.
Oncotarget ; 5(19): 8970-85, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25344860

ABSTRACT

Kindlin-3 (FERMT-3) is known to be central in hemostasis and thrombosis control and its deficiency disrupts platelet aggregation and causes Leukocyte Adhesion Deficiency disease. Here we report that Kindlin-3 has a tumor suppressive role in solid cancer. Our present genetic and functional data show that Kindlin-3 is downregulated in several solid tumors by a mechanism involving gene hypermethylation and deletions. In vivo experiments demonstrated that Kindlin-3 knockdown in 2 tumor cell models (breast cancer and melanoma) markedly increases metastasis formation, in accord with the in vitro increase of tumor cell malignant properties. The metastatic phenotype was supported by a mechanism involving alteration in ß3-integrin activation including decreased phosphorylation, interaction with talin and the internalization of its active form leading to less cell attachment and more migration/invasion. These data uncover a novel and unexpected tumor suppressor role of Kindlin-3 which can influence integrins targeted therapies development.


Subject(s)
Breast Neoplasms/pathology , Genes, Tumor Suppressor , Melanoma/pathology , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DNA Methylation , Decitabine , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Integrin beta3/metabolism , Melanoma/drug therapy , Melanoma/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Phosphorylation , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Talin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...