Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Environ Monit Assess ; 195(1): 147, 2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36422741

ABSTRACT

The Yaqui River Irrigation District is a region in Mexico with intensive agricultural production; thus, large quantities of fertilizers are used, and excess fertilizer can affect the quality of water bodies. The aim of this work was to estimate the water budget and nitrogen (N) and phosphorous (P) mass budgets to evaluate possible contamination of a littoral water body (Tóbari Bay) and leachates into an aquifer (Yaqui Valley aquifer). Wheat and corn crops were studied, climate information was compiled, and soil and water samples were collected for analysis. The water budget showed excess irrigation occurred due to the need for soil washing to prevent salinization. A total of 24% of all irrigation water was used for crops, 60% was discharged into the bay through runoff of the drainage system, and 16% corresponded to effective infiltration (aquifer recharge). The N budget showed that of the 100% N input, the highest percentage was used by plants (63%), and only minimal loss occurred through runoff (11%) and leachate into the aquifer (7%). The remaining N stayed in the soil (18%) or was volatilized (1%). These results indicate that the drainage system prevented large amounts of N from entering the aquifer; thus, the N concentrations in the groundwater did not exceed the regulated maximum limit for drinking water (10 mg N-NO3/L). In terms of the water pollution level in the bay, the presence of NO3- was minimal (concentrations below the quasintifiable limit). Of the 100% of P that was applied, 55% was used by the plants, and 40% remained in the soil; therefore, the P that was transported by runoff or was leached was minimal (3 and 2%, respectively). However, this minimal amount of P ranged from 0.1 to 0.2 mg/L in the bay, and these values exceeded the suggested values for the protection of aquatic life (0.01 mg/L). The administrators of the irrigation district must pay special attention to phosphate fertilizer management and implement irrigation techniques that increase water use efficiency.


Subject(s)
Drinking Water , Groundwater , Phosphorus/analysis , Nitrogen/analysis , Fertilizers/analysis , Mexico , Environmental Monitoring/methods , Groundwater/analysis , Drinking Water/analysis , Crops, Agricultural , Soil
2.
Sci Total Environ ; 648: 530-541, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30125849

ABSTRACT

Golf courses represent an agricultural activity wherein grass is intensively cultivated using large quantities of fertilizers. In the present study, nitrogen and phosphorus leaching was analyzed over two years in an experimental green under actual field conditions. The green contained four plots with distinct amendments (P1: hydrogel + peat, P2: peat, P3: hydrogel, and P4: no amendment). The applied doses of nitrogen ranged from 5 to 103 kg/ha and of phosphorus from 9 to 31 kg/ha. The irrigation level varied as a function of the rainfall regime and the water requirements of grass; overall water intake varied from 1550 to 2080 mm/year. Daily, leached water volume was calculated, and samples were taken for chemical analysis. Nitrogen and phosphorus mass balances were calculated for different periods based on the collected data. The plot amended with peat and hydrogel (P1) had reduced water flow; the percentage of drainage water varied from 8.4 to 29%. As a result, the dissolution and leaching of nitrogen (N) and phosphorus (P) were the lowest in comparison to the other plots. According to the calculated mass balances, the lowest leaching values were also recorded in this plot (P1), ranging from 0.5 to 6.3% for N and from 0.8 to 20.9% for P. The plot without amendment (P4) drained the most water (25.9-44.8%) and leached the highest quantities of N and P, ranging from 9.1-45.7%, and 6-35.9%, respectively. The use of double amendments (hydrogel and peat) therefore represented optimal operating conditions for the green. Moreover, a relationship was found between increasing rates of fertilization and increasing percentages of N and P leaching as well as between higher irrigation levels and greater leaching.

3.
Oncogene ; 38(5): 765-766, 2019 01.
Article in English | MEDLINE | ID: mdl-30397238

ABSTRACT

In the original article the authors have noted that the wrong image was used to illustrate the Uc.346 + Lu1-Lu2-Lu3 subpanel of Figure 5a. The correct image is now provided as Figure 1 in this article. This change does not affect the legend of the figure, the results, or conclusions reported in the manuscript. The authors apologize for the error, and regret any inconvenience this may have caused.

4.
Prog Neurobiol ; 168: 42-68, 2018 09.
Article in English | MEDLINE | ID: mdl-29653249

ABSTRACT

Human pluripotent stem cell (PSC) technology and direct somatic cell reprogramming have opened up a promising new avenue in the field of neuroscience. These recent advances allow researchers to obtain virtually any cell type found in the human brain, making it possible to produce and study functional neurons in laboratory conditions for both scientific and medical purposes. Although distinct approaches have shown to be successful in directing neuronal cell fate in vitro, their refinement and optimization, as well as the search for alternative approaches, remains necessary to help realize the full potential of the eventually derived neuronal populations. Furthermore, we are currently limited in the number of neuronal subtypes whose induction is fully established, and different cultivation protocols for each subtype exist, making it challenging to increase the reproducibility and decrease the variances that are observed between different protocols. In this review, we summarize the progress that has been made in generating various neuronal subtypes from PSCs and somatic cells, with special emphasis on chemically defined systems, transcription factor-mediated reprogramming and epigenetic-based approaches. We also discuss the efforts that are being made to increase the efficiency of current protocols and address the potential for the use of these cells in disease modelling, drug discovery and regenerative medicine.


Subject(s)
Cellular Reprogramming/physiology , Neurons/physiology , Pluripotent Stem Cells/physiology , Pluripotent Stem Cells/transplantation , Animals , Cell Differentiation/drug effects , Epigenomics/methods , In Vitro Techniques , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Oncogenesis ; 6(10): e390, 2017 Oct 23.
Article in English | MEDLINE | ID: mdl-29058695

ABSTRACT

Breast cancer-associated fibroblasts (CAFs) have a crucial role in tumor initiation, metastasis and therapeutic resistance by secreting various growth factors, cytokines, protease and extracellular matrix components. Soluble factors secreted by CAFs are involved in many pathways including inflammation, metabolism, proliferation and epigenetic modulation, suggesting that CAF-dependent reprograming of cancer cells affects a large set of genes. This paracrine signaling has an important role in tumor progression, thus deciphering some of these processes could lead to relevant discoveries with subsequent clinical implications. Here, we investigated the mechanisms underlying the changes in gene expression patterns associated with the cross-talk between breast cancer cells and the stroma. From RNAseq data obtained from breast cancer cell lines grown in presence of CAF-secreted factors, we identified 372 upregulated genes, exhibiting an expression level positively correlated with the stromal content of breast cancer specimens. Furthermore, we observed that gene expression changes were not mediated through significant DNA methylation changes. Nevertheless, CAF-secreted factors but also stromal content of the tumors remarkably activated specific genes characterized by a DNA methylation pattern: hypermethylation at transcription start site and shore regions. Experimental approaches (inhibition of DNA methylation, knockdown of methyl-CpG-binding domain protein 2 and chromatin immunoprecipitation assays) indicated that this set of genes was epigenetically controlled. These data elucidate the importance of epigenetics marks in the cancer cell reprogramming induced by stromal cell and indicated that the interpreters of the DNA methylation signal have a major role in the response of the cancer cells to the microenvironment.

6.
Oncogene ; 36(40): 5648-5657, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28581523

ABSTRACT

Although single base-pair resolution DNA methylation landscapes for embryonic and different somatic cell types provided important insights into epigenetic dynamics and cell-type specificity, such comprehensive profiling is incomplete across human cancer types. This prompted us to perform genome-wide DNA methylation profiling of 22 samples derived from normal tissues and associated neoplasms, including primary tumors and cancer cell lines. Unlike their invariant normal counterparts, cancer samples exhibited highly variable CpG methylation levels in a large proportion of the genome, involving progressive changes during tumor evolution. The whole-genome sequencing results from selected samples were replicated in a large cohort of 1112 primary tumors of various cancer types using genome-scale DNA methylation analysis. Specifically, we determined DNA hypermethylation of promoters and enhancers regulating tumor-suppressor genes, with potential cancer-driving effects. DNA hypermethylation events showed evidence of positive selection, mutual exclusivity and tissue specificity, suggesting their active participation in neoplastic transformation. Our data highlight the extensive changes in DNA methylation that occur in cancer onset, progression and dissemination.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Neoplasms/genetics , Animals , Base Pairing , Enhancer Elements, Genetic , Genome, Human , Humans , Promoter Regions, Genetic
9.
Sci Rep ; 7: 41903, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28211912

ABSTRACT

The characterization of the epigenetic changes within the obesity-related adipose tissue will provide new insights to understand this metabolic disorder, but adipose tissue is not easy to sample in population-based studies. We aimed to evaluate the capacity of circulating leukocytes to reflect the adipose tissue-specific DNA methylation status of obesity susceptibility. DNA samples isolated from subcutaneous adipose tissue and circulating leukocytes were hybridized in the Infinium HumanMethylation 450 BeadChip. Data were compared between samples from obese (n = 45) and non-obese (n = 8-10) patients by Wilcoxon-rank test, unadjusted for cell type distributions. A global hypomethylation of the differentially methylated CpG sites (DMCpGs) was observed in the obese subcutaneous adipose tissue and leukocytes. The overlap analysis yielded a number of genes mapped by the common DMCpGs that were identified to reflect the obesity state in the leukocytes. Specifically, the methylation levels of FGFRL1, NCAPH2, PNKD and SMAD3 exhibited excellent and statistically significant efficiencies in the discrimination of obesity from non-obesity status (AUC > 0.80; p < 0.05) and a great correlation between both tissues. Therefore, the current study provided new and valuable DNA methylation biomarkers of obesity-related adipose tissue pathogenesis through peripheral blood analysis, an easily accessible and minimally invasive biological material instead of adipose tissue.


Subject(s)
DNA Methylation , Leukocytes/metabolism , Obesity/genetics , Subcutaneous Fat/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , CpG Islands , Female , Genome, Human , Humans , Male , Middle Aged , Muscle Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 5/genetics , Serine Endopeptidases/genetics , Smad3 Protein/genetics
10.
Mol Psychiatry ; 22(4): 485-496, 2017 04.
Article in English | MEDLINE | ID: mdl-28044062

ABSTRACT

Dementia is a complex clinical condition characterized by several cognitive impairments that interfere with patient independence in executing everyday tasks. Various neurodegenerative disorders have dementia in common among their clinical manifestations. In addition, these diseases, such as Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies and frontotemporal dementia, share molecular alterations at the neuropathological level. In recent years, the field of neuroepigenetics has expanded massively and it is now clear that epigenetic processes, such as DNA methylation, are mechanisms involved in both normal and pathological brain function. Despite the persistent methodological and conceptual caveats, it has been reported that several genes fundamental to the development of neurodegenerative disorders are deregulated by aberrant methylation patterns of their promoters, and even common epigenetic signatures for some dementia-associated pathologies have been identified. Therefore, understanding the epigenetic mechanisms that are altered in dementia, especially those associated with the initial phases, will allow us not only to understand the etiopathology of dementia and its progression but also to design effective therapies to reduce this global public health problem. This review provides an in-depth summary of our current knowledge about DNA methylation in dementia, focusing exclusively on the analyses performed in human brain.


Subject(s)
Dementia/metabolism , Dementia/physiopathology , Epigenesis, Genetic/physiology , Alzheimer Disease/genetics , Brain/pathology , Cognition Disorders/genetics , DNA/genetics , DNA Methylation/physiology , Epigenesis, Genetic/genetics , Epigenomics/methods , Humans , Neurodegenerative Diseases/genetics , Parkinson Disease/genetics
11.
Oncogene ; 36(9): 1287-1296, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27593925

ABSTRACT

Components of the SWI/SNF chromatin remodeling complex, including BRG1 (also SMARCA4), are inactivated in cancer. Among other functions, SWI/SNF orchestrates the response to retinoid acid (RA) and glucocorticoids (GC) involving downregulation of MYC. The epigenetic drugs SAHA and azacytidine, as well as RA and GC, are currently being used to treat some malignancies but their therapeutic potential in lung cancer is not well established. Here we aimed to determine the possible therapeutic effects of azacytidine and SAHA (A/S) alone or in combination with GC plus RA (GC/RA) in lung cancers with either BRG1 inactivation or MYC amplification. In vitro, responses to GC/RA treatment were more effective in MYC-amplified cells. These effects were mediated by BRG1 and involved a reprogramming towards prodifferentiation gene expression signatures and downregulation of MYC. In MYC-amplified cells, administration of GC/RA enhanced the cell growth inhibitory effects of A/S which, in turn, accentuated the prodifferentiation features promoted by GC/RA. Finally, these treatments improved overall survival of mice orthotopically implanted with MYC-amplified, but not BRG1-mutant, cells and reduced tumor cell viability and proliferation. We propose that the combination of epigenetic treatments with retinoids and corticoids of MYC-driven lung tumors constitute a strategy for therapeutic intervention in this otherwise incurable disease.


Subject(s)
Azacitidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Glucocorticoids/pharmacology , Hydroxamic Acids/pharmacology , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , Tretinoin/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , DNA Helicases/genetics , DNA Methylation , Histone Deacetylase Inhibitors/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Mutation/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Tumor Cells, Cultured , Vorinostat , Xenograft Model Antitumor Assays
12.
Transl Res ; 178: 13-24.e5, 2016 12.
Article in English | MEDLINE | ID: mdl-27477082

ABSTRACT

Elucidating the potential mechanisms involved in the detrimental effect of excess body weight on insulin action is an important priority in counteracting obesity-associated diseases. The present study aimed to disentangle the epigenetic basis of insulin resistance by performing a genome-wide epigenetic analysis in visceral adipose tissue (VAT) from morbidly obese patients depending on the insulin sensitivity evaluated by the clamp technique. The global human methylome screening performed in VAT from 7 insulin-resistant (IR) and 5 insulin-sensitive (IS) morbidly obese patients (discovery cohort) analyzed using the Infinium HumanMethylation450 BeadChip array identified 982 CpG sites able to perfectly separate the IR and IS samples. The identified sites represented 538 unique genes, 10% of which were diabetes-associated genes. The current work identified novel IR-related genes epigenetically regulated in VAT, such as COL9A1, COL11A2, CD44, MUC4, ADAM2, IGF2BP1, GATA4, TET1, ZNF714, ADCY9, TBX5, and HDACM. The gene with the largest methylation fold-change and mapped by 5 differentially methylated CpG sites located in island/shore and promoter region was ZNF714. This gene presented lower methylation levels in IR than in IS patients in association with increased transcription levels, as further reflected in a validation cohort (n = 24; 11 IR and 13 IS). This study reveals, for the first time, a potential epigenetic regulation involved in the dysregulation of VAT that could predispose patients to insulin resistance and future type 2 diabetes in morbid obesity, providing a potential therapeutic target and biomarkers for counteracting this process.


Subject(s)
DNA Methylation/genetics , Genome, Human , Insulin Resistance/genetics , Insulin/pharmacology , Intra-Abdominal Fat/metabolism , Obesity, Morbid/genetics , Anthropometry , Chromosomes, Human/genetics , Cohort Studies , CpG Islands/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Intra-Abdominal Fat/drug effects , Male , Middle Aged , Reproducibility of Results , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Ann Oncol ; 27(6): 1062-1067, 2016 06.
Article in English | MEDLINE | ID: mdl-26916096

ABSTRACT

BACKGROUND: Objective response to dacarbazine, the intravenous form of temozolomide (TMZ), in metastatic colorectal cancer (mCRC) is confined to tumors harboring O(6)-methylguanine-DNA-methyltransferase (MGMT) promoter hypermethylation. We conducted a phase II study of TMZ enriched by MGMT hypermethylation in archival tumor (AT), exploring dynamic of this biomarker in baseline tumor (BT) biopsy and plasma (liquid biopsy). PATIENTS AND METHODS: We screened 150 mCRC patients for MGMT hypermethylation with methylation-specific PCR on AT from FFPE specimens. Eligible patients (n = 29) underwent BT biopsy and then received TMZ 200 mg/m(2) days 1-5 q28 until progression. A Fleming single-stage design was used to determine whether progression-free survival (PFS) rate at 12 weeks would be ≥35% [H0 ≤ 15%, type I error = 0.059 (one-sided), power = 0.849]. Exploratory analyses included comparison between MGMT hypermethylation in AT and BT, and MGMT methylation testing by MethylBEAMing in solid (AT, BT) and LB with regard to tumor response. RESULTS: The PFS rate at 12 weeks was 10.3% [90% confidence interval (CI) 2.9-24.6]. Objective response rate was 3.4% (90% CI 0.2-15.3), disease control rate 48.3% (90% CI 32.0-64.8), median OS 6.2 months (95% CI 3.8-7.6), and median PFS 2.6 months (95% CI 1.4-2.7). We observed the absence of MGMT hypermethylation in BT in 62.7% of tumors. CONCLUSION: Treatment of mCRC with TMZ driven by MGMT promoter hypermethylation in AT samples did not provide meaningful PFS rate at 12 weeks. This biomarker changed from AT to BT, indicating that testing BT biopsy or plasma is needed for refined target selection.


Subject(s)
Colorectal Neoplasms/drug therapy , DNA Methylation/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Tumor Suppressor Proteins/genetics , Adult , Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Biopsy , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Modification Methylases/blood , DNA Repair Enzymes/blood , Dacarbazine/administration & dosage , Disease-Free Survival , Female , Humans , Male , Middle Aged , Promoter Regions, Genetic , Temozolomide , Tumor Suppressor Proteins/blood
15.
Transl Psychiatry ; 6: e718, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26784972

ABSTRACT

Different neurodegenerative disorders often show similar lesions, such as the presence of amyloid plaques, TAU-neurotangles and synuclein inclusions. The genetically inherited forms are rare, so we wondered whether shared epigenetic aberrations, such as those affecting DNA methylation, might also exist. The studied samples were gray matter samples from the prefrontal cortex of control and neurodegenerative disease-associated cases. We performed the DNA methylation analyses of Alzheimer's disease, dementia with Lewy bodies, Parkinson's disease and Alzheimer-like neurodegenerative profile associated with Down's syndrome samples. The DNA methylation landscapes obtained show that neurodegenerative diseases share similar aberrant CpG methylation shifts targeting a defined gene set. Our findings suggest that neurodegenerative disorders might have similar pathogenetic mechanisms that subsequently evolve into different clinical entities. The identified aberrant DNA methylation changes can be used as biomarkers of the disorders and as potential new targets for the development of new therapies.


Subject(s)
DNA Methylation/physiology , Epigenomics , Neurodegenerative Diseases/metabolism , Prefrontal Cortex/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Tissue Array Analysis
16.
Oncogene ; 35(23): 3079-82, 2016 06 09.
Article in English | MEDLINE | ID: mdl-26434589

ABSTRACT

Close to half of de novo acute myeloid leukemia (AML) cases do not exhibit any cytogenetic aberrations. In this regard, distortion of the DNA methylation setting and the presence of mutations in epigenetic modifier genes can also be molecular drivers of the disease. In recent years, somatic missense mutations of the DNA methyltransferase 3A (DNMT3A) have been reported in ~20% of AML patients; however, no obvious critical downstream gene has been identified that could explain the role of DNMT3A in the natural history of AML. Herein, using whole-genome bisulfite sequencing and DNA methylation microarrays, we have identified a key gene undergoing promoter hypomethylation-associated transcriptional reactivation in DNMT3 mutant patients, the leukemogenic HOX cofactor MEIS1. Our results indicate that, in the absence of mixed lineage leukemia fusions, an alternative pathway for engaging an oncogenic MEIS1-dependent transcriptional program can be mediated by DNMT3A mutations.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Neoplasm Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism
17.
Oncogene ; 35(33): 4407-13, 2016 08 18.
Article in English | MEDLINE | ID: mdl-26640150

ABSTRACT

The introduction of new therapies against particular genetic mutations in non-small-cell lung cancer is a promising avenue for improving patient survival, but the target population is small. There is a need to discover new potential actionable genetic lesions, to which end, non-conventional cancer pathways, such as RNA editing, are worth exploring. Herein we show that the adenosine-to-inosine editing enzyme ADAR1 undergoes gene amplification in non-small cancer cell lines and primary tumors in association with higher levels of the corresponding mRNA and protein. From a growth and invasion standpoint, the depletion of ADAR1 expression in amplified cells reduces their tumorigenic potential in cell culture and mouse models, whereas its overexpression has the opposite effects. From a functional perspective, ADAR1 overexpression enhances the editing frequencies of target transcripts such as NEIL1 and miR-381. In the clinical setting, patients with early-stage lung cancer, but harboring ADAR1 gene amplification, have poor outcomes. Overall, our results indicate a role for ADAR1 as a lung cancer oncogene undergoing gene amplification-associated activation that affects downstream RNA editing patterns and patient prognosis.


Subject(s)
Adenosine Deaminase/genetics , Gene Amplification , Lung Neoplasms/etiology , RNA Editing , RNA-Binding Proteins/genetics , Cell Line, Tumor , Humans , Lung Neoplasms/genetics , Oncogenes , Proto-Oncogene Proteins p21(ras)/genetics
18.
PLoS One ; 10(7): e0132517, 2015.
Article in English | MEDLINE | ID: mdl-26161907

ABSTRACT

INTRODUCTION AND RESULTS: Immunodeficiency, centromeric instability and facial anomalies syndrome (ICF) is a rare autosomal recessive disease, characterized by severe hypomethylation in pericentromeric regions of chromosomes (1, 16 and 9), marked immunodeficiency and facial anomalies. The majority of ICF patients present mutations in the DNMT3B gene, affecting the DNA methyltransferase activity of the protein. In the present study, we have used the Infinium 450K DNA methylation array to evaluate the methylation level of 450,000 CpGs in lymphoblastoid cell lines and untrasformed fibroblasts derived from ICF patients and healthy donors. Our results demonstrate that ICF-specific DNMT3B variants A603T/STP807ins and V699G/R54X cause global DNA hypomethylation compared to wild-type protein. We identified 181 novel differentially methylated positions (DMPs) including subtelomeric and intrachromosomic regions, outside the classical ICF-related pericentromeric hypomethylated positions. Interestingly, these sites were mainly located in intergenic regions and inside the CpG islands. Among the identified hypomethylated CpG-island associated genes, we confirmed the overexpression of three selected genes, BOLL, SYCP2 and NCRNA00221, in ICF compared to healthy controls, which are supposed to be expressed in germ line and silenced in somatic tissues. CONCLUSIONS: In conclusion, this study contributes in clarifying the direct relationship between DNA methylation defect and gene expression impairment in ICF syndrome, identifying novel direct target genes of DNMT3B. A high percentage of the DMPs are located in the subtelomeric regions, indicating a specific role of DNMT3B in methylating these chromosomal sites. Therefore, we provide further evidence that hypomethylation in specific non-pericentromeric regions of chromosomes might be involved in the molecular pathogenesis of ICF syndrome. The detection of DNA hypomethylation at BOLL, SYCP2 and NCRNA00221 may pave the way for the development of specific clinical biomarkers with the aim to facilitate the identification of ICF patients.


Subject(s)
Centromere/genetics , DNA Methylation/genetics , Genome, Human , Immunologic Deficiency Syndromes/genetics , Case-Control Studies , Cell Line , Chromosomes, Human/genetics , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Female , Gene Expression Regulation , Genetic Variation , Humans , Male , Promoter Regions, Genetic/genetics , Reproducibility of Results , Sequence Analysis, DNA , DNA Methyltransferase 3B
19.
Ann Oncol ; 26(9): 1994-1999, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26113646

ABSTRACT

BACKGROUND: O(6)-methyl-guanine-methyl-transferase (MGMT) silencing by promoter methylation may identify cancer patients responding to the alkylating agents dacarbazine or temozolomide. PATIENTS AND METHODS: We evaluated the prognostic and predictive value of MGMT methylation testing both in tumor and cell-free circulating DNA (cfDNA) from plasma samples using an ultra-sensitive two-step digital PCR technique (methyl-BEAMing). Results were compared with two established techniques, methylation-specific PCR (MSP) and Bs-pyrosequencing. RESULTS: Thresholds for MGMT methylated status for each technique were established in a training set of 98 glioblastoma (GBM) patients. The prognostic and the predictive value of MGMT methylated status was validated in a second cohort of 66 GBM patients treated with temozolomide in which methyl-BEAMing displayed a better specificity than the other techniques. Cutoff values of MGMT methylation specific for metastatic colorectal cancer (mCRC) tissue samples were established in a cohort of 60 patients treated with dacarbazine. In mCRC, both quantitative assays methyl-BEAMing and Bs-pyrosequencing outperformed MSP, providing better prediction of treatment response and improvement in progression-free survival (PFS) (P < 0.001). Ability of methyl-BEAMing to identify responding patients was validated in a cohort of 23 mCRC patients treated with temozolomide and preselected for MGMT methylated status according to MSP. In mCRC patients treated with dacarbazine, exploratory analysis of cfDNA by methyl-BEAMing showed that MGMT methylation was associated with better response and improved median PFS (P = 0.008). CONCLUSIONS: Methyl-BEAMing showed high reproducibility, specificity and sensitivity and was applicable to formalin-fixed paraffin-embedded tissues and cfDNA. This study supports the quantitative assessment of MGMT methylation for clinical purposes since it could refine prediction of response to alkylating agents.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , DNA Methylation/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Glioblastoma/drug therapy , Tumor Suppressor Proteins/metabolism , Brain Neoplasms/mortality , Colorectal Neoplasms/mortality , DNA/blood , DNA/metabolism , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Disease-Free Survival , Glioblastoma/mortality , Humans , Polymerase Chain Reaction , Prognosis , Promoter Regions, Genetic/genetics , Temozolomide , Tumor Suppressor Proteins/genetics
20.
Cell Death Dis ; 6: e1635, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25675295

ABSTRACT

The generation of B cells is a complex process requiring several cellular transitions, including cell commitment and differentiation. Proper transcriptional control to establish the genetic programs characteristic of each cellular stage is essential for the correct development of B lymphocytes. Deregulation of these particular transcriptional programs may result in a block in B-cell maturation, contributing to the development of hematological malignancies such as leukemia and lymphoma. However, very little is currently known about the role of transcriptional repressors in normal and aberrant B lymphopoiesis. Here we report that histone deacetylase 7 (HDAC7) is underexpressed in pro-B acute lymphoblastic leukemia (pro-B-ALL) and Burkitt lymphoma. Ectopic expression of HDAC7 induces apoptosis, leads to the downregulation of c-Myc and inhibits the oncogenic potential of cells in vivo, in a xenograft model. Most significantly, we have observed low levels of HDAC7 expression in B-ALL patient samples, which is correlated with the increased levels of c-Myc. From a mechanistic angle, we show that ectopically expressed HDAC7 localizes to the nucleus and interacts with the transcription factor myocyte enhancer factor C (MEF2C) and the corepressors HDAC3 and SMRT. Accordingly, both the HDAC7-MEF2C interaction domain as well as its catalytic domain are involved in the reduced cell viability induced by HDAC7. We conclude that HDAC7 has a potent anti-oncogenic effect on specific B-cell malignancies, indicating that its deregulation may contribute to the pathogenesis of the disease.


Subject(s)
Down-Regulation/genetics , Histone Deacetylases/metabolism , Leukemia/metabolism , Lymphoma/metabolism , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis/genetics , Apoptosis/physiology , B-Lymphocytes/metabolism , Cell Cycle , Cell Line, Tumor , Cell Nucleus/metabolism , Cellular Reprogramming Techniques , Histone Deacetylases/genetics , Humans , MEF2 Transcription Factors/metabolism , Male , Mice , Nuclear Receptor Co-Repressor 2 , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...