Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Ageing Res Rev ; 37: 28-38, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28499883

ABSTRACT

DNA methylation at the fifth position of cytosines (5mC) represents a major epigenetic modification in mammals. The recent discovery of 5-hydroxymethylcytosine (5hmC), resulting from 5mC oxidation, is redefining our view of the epigenome, as multiple studies indicate that 5hmC is not simply an intermediate of DNA demethylation, but a genuine epigenetic mark that may play an important functional role in gene regulation. Currently, the availability of platforms that discriminates between the presence of 5mC and 5hmC at single-base resolution is starting to shed light on the functions of 5hmC. In this review, we provide an overview of the genomic distribution of 5hmC, and examine recent findings on the role of this mark and the potential consequences of its misregulation during three fundamental biological processes: cell differentiation, cancer and aging.


Subject(s)
5-Methylcytosine/analogs & derivatives , Aging/metabolism , DNA Methylation , Epigenesis, Genetic , Neoplasms/metabolism , 5-Methylcytosine/metabolism , Animals , Cell Differentiation , Cytosine , Humans , Neoplasms/genetics , Oxidation-Reduction
2.
Int J Nanomedicine ; 11: 6297-6306, 2016.
Article in English | MEDLINE | ID: mdl-27932878

ABSTRACT

Human exposure to nanomaterials and nanoparticles is increasing rapidly, but their effects on human health are still largely unknown. Epigenetic modifications are attracting ever more interest as possible underlying molecular mechanisms of gene-environment interactions, highlighting them as potential molecular targets following exposure to nanomaterials and nanoparticles. Interestingly, recent research has identified changes in DNA methylation, histone post-translational modifications, and noncoding RNAs in mammalian cells exposed to nanomaterials and nanoparticles. However, the challenge for the future will be to determine the molecular pathways driving these epigenetic alterations, the possible functional consequences, and the potential effects on health.


Subject(s)
Environmental Exposure/adverse effects , Epigenesis, Genetic/drug effects , Mammals/genetics , Nanoparticles/toxicity , Animals , DNA Methylation/drug effects , DNA Methylation/genetics , Histones/metabolism , Humans , Protein Processing, Post-Translational/drug effects
3.
J Mol Neurosci ; 58(2): 243-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26553261

ABSTRACT

The liver X receptor agonist, GW3965, improves cognition in Alzheimer's disease (AD) mouse models. Here, we determined if short-term GW3965 treatment induces changes in the DNA methylation state of the hippocampus, which are associated with cognitive improvement. Twenty-four-month-old triple-transgenic AD (3xTg-AD) mice were treated with GW3965 (50 mg/kg/day for 6 days). DNA methylation state was examined by modified bisulfite conversion and hybridization on Illumina Infinium Methylation BeadChip 450 k arrays. The Morris water maze was used for behavioral analysis. Our results show in addition to improvement in cognition methylation changes in 39 of 13,715 interrogated probes in treated 3xTg-AD mice compared with untreated 3xTg-AD mice. These changes in methylation probes include 29 gene loci. Importantly, changes in methylation status were mainly from synapse-related genes (SYP, SYN1, and DLG3) and neurogenesis-associated genes (HMGB3 and RBBP7). Thus, our results indicate that liver X receptors (LXR) agonist treatment induces rapid changes in DNA methylation, particularly in loci associated with genes involved in neurogenesis and synaptic function. Our results suggest a new potential mechanism to explain the beneficial effect of GW3965.


Subject(s)
Alzheimer Disease/metabolism , Benzoates/pharmacology , Benzylamines/pharmacology , DNA Methylation/drug effects , Neurogenesis , Orphan Nuclear Receptors/agonists , Synapses/drug effects , Alzheimer Disease/genetics , Animals , Female , HMGB3 Protein/genetics , HMGB3 Protein/metabolism , Liver X Receptors , Mice , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Retinoblastoma-Binding Protein 7/genetics , Retinoblastoma-Binding Protein 7/metabolism , Synapses/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism
4.
Leukemia ; 30(3): 674-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26500142

ABSTRACT

B cells have been shown to be refractory to reprogramming and B-cell-derived induced pluripotent stem cells (iPSC) have only been generated from murine B cells engineered to carry doxycycline-inducible Oct4, Sox2, Klf4 and Myc (OSKM) cassette in every tissue and from EBV/SV40LT-immortalized lymphoblastoid cell lines. Here, we show for the first time that freshly isolated non-cultured human cord blood (CB)- and peripheral blood (PB)-derived CD19+CD20+ B cells can be reprogrammed to iPSCs carrying complete VDJH immunoglobulin (Ig) gene monoclonal rearrangements using non-integrative tetracistronic, but not monocistronic, OSKM-expressing Sendai Virus. Co-expression of C/EBPα with OSKM facilitates iPSC generation from both CB- and PB-derived B cells. We also demonstrate that myeloid cells are much easier to reprogram than B and T lymphocytes. Differentiation potential back into the cell type of their origin of B-cell-, T-cell-, myeloid- and fibroblast-iPSCs is not skewed, suggesting that their differentiation does not seem influenced by 'epigenetic memory'. Our data reflect the actual cell-autonomous reprogramming capacity of human primary B cells because biased reprogramming was avoided by using freshly isolated primary cells, not exposed to cytokine cocktails favoring proliferation, differentiation or survival. The ability to reprogram CB/PB-derived primary human B cells offers an unprecedented opportunity for studying developmental B lymphopoiesis and modeling B-cell malignancies.


Subject(s)
B-Lymphocytes/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cellular Reprogramming/genetics , Fetal Blood/metabolism , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Base Sequence , CCAAT-Enhancer-Binding Proteins/immunology , Cell Differentiation , Cell Separation , Cellular Reprogramming/immunology , Fetal Blood/cytology , Fetal Blood/immunology , Gene Expression , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Molecular Sequence Data , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/immunology , Primary Cell Culture , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/immunology , Sendai virus/genetics , V(D)J Recombination/immunology
5.
Leukemia ; 30(1): 94-103, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26216197

ABSTRACT

The JAK-STAT pathway has a substantial role in lymphoid precursor cell proliferation, survival and differentiation. Nonetheless, the contribution of JAK2 to T-cell lymphoblastic lymphoma (T-LBL) development remains poorly understood. We have identified one activating TEL-JAK2 translocation and four missense mutations accumulated in 2 out of 16 T-LBL samples. Two of them are novel JAK2 mutations and the other two are reported for the first time in T-LBL. Notably, R683G and I682T might have arisen owing to RNA editing. Mutated samples showed different mutated transcripts suggesting sub-clonal heterogeneity. Functional approaches revealed that two JAK2 mutations (H574R and R683G) constitutively activate JAK-STAT signaling in γ2A cells and can drive the proliferation of BaF3-EpoR cytokine-dependent cell line. In addition, aberrant hypermethylation of SOCS3 might contribute to enhance the activation of JAK-STAT signaling. Of utmost interest is that primary T-LBL samples harboring JAK2 mutations exhibited increased expression of LMO2, suggesting a mechanistic link between JAK2 mutations and the expression of LMO2, which was confirmed for the four missense mutations in transfected γ2A cells. We therefore propose that active JAK2 contribute to T-LBL development by two different mechanisms, and that the use of pan-JAK inhibitors in combination with epigenetic drugs should be considered in future treatments.


Subject(s)
Janus Kinase 2/genetics , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adaptor Proteins, Signal Transducing/genetics , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 9 , DNA Methylation , Humans , Janus Kinase 2/physiology , LIM Domain Proteins/genetics , Proto-Oncogene Proteins/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Translocation, Genetic
6.
Genes Immun ; 16(1): 71-82, 2015.
Article in English | MEDLINE | ID: mdl-25393931

ABSTRACT

Engagement of the activating receptor NKG2D (natural killer group 2 member D) with its ligands (NKG2DL) major histocompatibility complex class I related-A and -B (MICA/B), UL-16 binding protein families (ULBPs 1-6) is important to ensure the innate immunity to tumor cells. However, these cells have developed strategies to downregulate NKG2DL expression and avoid immune recognition. We demonstrate that DNA methylation can contribute to the absence of NKG2DL expression during tumor progression. We analyzed the DNA methylation profiles for each NKG2DL by pyrosequencing in acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), hepatocellular carcinoma (HC), breast cancer and colon cancer cell lines. High levels of DNA methylation for NKG2DL were found in some tumor cell lines, mainly in AML cells. This hypermethylation was correlated with the absence of transcription for NKG2DL. Higher DNA methylation levels for MICA, ULBP1 and ULBP2 were observed in AML patients (n=60) compared with healthy donors (n=25). However, no DNA methylation for NKG2DL was found in colon cancer patients (n=44). Treatment with demethylating agents (5-azacytidine and 5-aza-2'-deoxycytidine) restored the expression of NKG2DL on the cell surface of AML cells, leading to an enhanced recognition by NKG2D-expressing cells. Our data suggest that NKG2DL may be aberrantly silenced by DNA methylation as a consequence of tumor development in AML patients.


Subject(s)
GPI-Linked Proteins/metabolism , Leukemia, Myeloid, Acute/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Tumor Escape , Cell Line, Tumor , DNA Methylation , GPI-Linked Proteins/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Ligands , NK Cell Lectin-Like Receptor Subfamily K/immunology
7.
Br J Cancer ; 110(8): 2123-30, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24595004

ABSTRACT

BACKGROUND: Aberrant global DNA methylation is shown to increase cancer risk. LINE-1 has been proven a measure of global DNA methylation. The objectives of this study were to assess the association between LINE-1 methylation level and bladder cancer risk and to evaluate effect modification by environmental and genetic factors. METHODS: Bisulphite-treated leukocyte DNA from 952 cases and 892 hospital controls was used to measure LINE-1 methylation level at four CpG sites by pyrosequencing. Logistic regression model was fitted to estimate odds ratios (ORs) and 95% confidence intervals (95% CIs). Interactions between LINE-1 methylation levels and environmental and genetic factors were assessed. RESULTS: The risk of bladder cancer followed a nonlinear association with LINE-1 methylation. Compared with subjects in the middle tertile, the adjusted OR for subjects in the lower and the higher tertiles were 1.26 (95% CI 0.99-1.60, P=0.06) and 1.33 (95% CI 1.05-1.69, P=0.02), respectively. This association significantly increased among individuals homozygous for the major allele of five single-nucleotide polymorphisms located in the phosphatidylethanolamine N-methyltransferase gene (corrected P-interaction<0.05). CONCLUSIONS: The findings from this large-scale study suggest that both low and high levels of global DNA methylation are associated with the risk of bladder cancer.


Subject(s)
DNA Methylation/genetics , Long Interspersed Nucleotide Elements/genetics , Phosphatidylethanolamine N-Methyltransferase/genetics , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , CpG Islands/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Leukocytes/pathology , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors , Urinary Bladder Neoplasms/pathology
8.
Clin Exp Immunol ; 176(2): 255-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24666184

ABSTRACT

Human adipose mesenchymal stem cells are a heterogeneous population, where cell cultures derived from single-cell-expanded clones present varying degrees of differential plasticity. This work focuses on the immunomodulatory/anti-inflammatory properties of these cells. To this end, five single-cell clones were isolated (generally called 1.X and 3.X) from two volunteers. Regarding the expression level of the lineage-characteristic surface antigens, clones 1·10 and 1·22 expressed the lowest amounts, while clones 3·10 and 3·5 expressed more CD105 than the rest and clone 1·7 expressed higher amounts of CD73 and CD44. Regarding cytokine secretion, all clones were capable of spontaneously releasing high levels of interleukin (IL)-6 and low to moderate levels of IL-8. These differences can be explained in part by the distinct methylation profile exhibited by the clones. Furthermore, and after lipopolysaccharide stimulation, clone 3.X produced the highest amounts of proinflammatory cytokines such as IL-1ß, while clones 1·10 and 1·22 highly expressed IL-4 and IL-5. In co-culture experiments, clones 1.X are, together, more potent inhibitors than clones 3.X for proliferation of total, CD3(+) T, CD4(+) T and CD8(+) T lymphocytes and natural killer (NK) cells. The results of this work indicate that the adipose stem cell population is heterogeneous in cytokine production profile, and that isolation, characterization and selection of the appropriate cell clone is a more exact method for the possible treatment of different patients or pathologies.


Subject(s)
Adipose Tissue/cytology , Cytokines/immunology , Inflammation Mediators/immunology , Mesenchymal Stem Cells/cytology , Adipose Tissue/immunology , Adipose Tissue/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Clone Cells/cytology , Clone Cells/immunology , Clone Cells/metabolism , Cluster Analysis , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , DNA Methylation , Flow Cytometry , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukin-5/genetics , Interleukin-5/immunology , Interleukin-5/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
9.
Genes Cancer ; 4(3-4): 105-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-24020001

ABSTRACT

Sirtuins play an essential role in the cellular response to environmental stress, promoting DNA repair, telomere stability, cell cycle arrest, cellular senescence, and apoptosis. Much attention has been given to the role of sirtuins in aging and cancer development; however, less is known about their role in stem cell regulation. This review focuses in this topic and discusses the possible implications in adult stem cell aging.

10.
Cell Death Dis ; 4: e570, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23559004

ABSTRACT

Lineage commitment during embryonic stem cell (ESC) differentiation is controlled not only by a gamut of transcription factors but also by epigenetic events, mainly histone deacetylation and promoter DNA methylation. The DNA demethylation agent 5'-aza-2'-deoxycytidine (AzadC) has been widely described as an effective promoter of cardiomyogenic differentiation in various stem cell types. However, its toxicity and instability complicate its use. Therefore, the purpose of this study was to examine the effects of zebularine (1-(ß-D-ribofuranosyl)-1,2-dihydropyrimidin-2-1), a stable and non-toxic DNA cytosine methylation inhibitor, on mouse ESC (mESC) differentiation. Herein, we report that treating embryoid bodies, generated from mESCs, with 30 µM zebularine for 7 days led to greater cell differentiation and induced the expression of several cardiac-specific markers that were detected using reverse transcription-polymerase chain reaction (RT-PCR), real-time PCR, immunostaining and flow cytometry. Zebularine enhanced the expression of cardiac markers and the appearance of beating cells that responded to cardiac drugs, including ion channel blockers (diltiazem) and ß-adrenergic stimulators (isoproterenol). Gene promoter methylation status was assessed using methylation-specific PCR (MSP) and validated by bisulfite sequencing analysis. Global gene expression profiling using microarrays showed that zebularine-differentiated cells are distinct from control ESCs. Pathway analysis revealed an enhancement of cellular processes such as embryonic development, cardiovascular system development and function. In addition, the whole-cell proteins exhibited different profiles as analyzed by two-dimensional differential-in-gel-electrophoresis. Our results indicate that zebularine regulates mesodermal differentiation of mESCs, controls promoter methylation of crucial cardiac genes and may help to improve cardiomyogenic differentiation.


Subject(s)
Cytidine/analogs & derivatives , Embryoid Bodies/drug effects , Metabolic Networks and Pathways/drug effects , Myocytes, Cardiac/drug effects , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line , Cytidine/pharmacology , DNA Methylation/drug effects , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Real-Time Polymerase Chain Reaction
11.
Diabetologia ; 55(12): 3341-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22961225

ABSTRACT

AIMS/HYPOTHESIS: Energy-dense diets that are high in fat are associated with a risk of metabolic diseases. The underlying molecular mechanisms could involve epigenetics, as recent data show altered DNA methylation of putative type 2 diabetes candidate genes in response to high-fat diets. We examined the effect of a short-term high-fat overfeeding (HFO) diet on genome-wide DNA methylation patterns in human skeletal muscle. METHODS: Skeletal muscle biopsies were obtained from 21 healthy young men after ingestion of a short-term HFO diet and a control diet, in a randomised crossover setting. DNA methylation was measured in 27,578 CpG sites/14,475 genes using Illumina's Infinium Bead Array. Candidate gene expression was determined by quantitative real-time PCR. RESULTS: HFO introduced widespread DNA methylation changes affecting 6,508 genes (45%), with a maximum methylation change of 13.0 percentage points. The HFO-induced methylation changes were only partly and non-significantly reversed after 6-8 weeks. Alterations in DNA methylation levels primarily affected genes involved in inflammation, the reproductive system and cancer. Few gene expression changes were observed and these had poor correlation to DNA methylation. CONCLUSIONS/INTERPRETATION: The genome-wide DNA methylation changes induced by the short-term HFO diet could have implications for our understanding of transient epigenetic regulation in humans and its contribution to the development of metabolic diseases. The slow reversibility suggests a methylation build-up with HFO, which over time may influence gene expression levels.


Subject(s)
DNA Methylation , Diet, High-Fat , Muscle, Skeletal/metabolism , Cation Transport Proteins/genetics , CpG Islands/genetics , Cross-Over Studies , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Gene Expression , Heat-Shock Proteins/genetics , Homeodomain Proteins/genetics , Humans , Insulin Resistance/genetics , Male , Muscle, Skeletal/physiology , Overnutrition , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Proto-Oncogene Proteins c-akt/genetics , Real-Time Polymerase Chain Reaction , Trans-Activators/genetics , Transcription Factors/genetics , Young Adult , Zinc Transporter 8
12.
J Mol Med (Berl) ; 90(5): 587-95, 2012 May.
Article in English | MEDLINE | ID: mdl-22120039

ABSTRACT

The bromodomain protein BRD4 is involved in cell proliferation and cell cycle progression, primarily through its role in acetylated chromatin-dependent regulation of transcription at targeted loci. Here, we show that BRD4 is frequently downregulated by aberrant promoter hypermethylation in human colon cancer cell lines and primary tumors. Ectopic re-expression of BRD4 in these colon cancer cell lines markedly reduced in vivo tumor growth, suggesting a role of BRD4 in human colon cancer.


Subject(s)
Colonic Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Transcription Factors/genetics , Acetylation , Animals , Cell Cycle Proteins , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA Methylation/genetics , Gene Silencing , Histones/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism
13.
Tissue Antigens ; 76(6): 431-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21058938

ABSTRACT

Epigenetics comprises various mechanisms that mold chromatin structures and regulate gene expression with stability, thus defining cell identity and function and adapting cells to environmental changes. Alteration of these mechanisms contributes to the inception of various pathological conditions. Given the complexity of the immune system, one would predict that a higher-order, supragenetic regulation is indispensable for generation of its constituents and control of its functions. Here, we summarize various aspects of immune system physiology and pathology in which epigenetic pathways have been implicated. Increasing knowledge in this field, together with the development of specific tools with which to manipulate epigenetic pathways, might form a basis for new strategies of immune function modulation, both to optimize immune therapies for infections or cancer and to control immune alterations in aging or autoimmunity.


Subject(s)
Autoimmunity , Epigenesis, Genetic/immunology , Immune System Diseases/immunology , Infections/immunology , Neoplasms/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Epigenesis, Genetic/genetics , Humans , Immune System Diseases/genetics , Infections/genetics , Neoplasms/genetics
14.
Obstet Gynecol Int ; 2010: 605163, 2010.
Article in English | MEDLINE | ID: mdl-21052542

ABSTRACT

Gestational diabetes mellitus (GDM) is defined as the glucose intolerance that is not present or recognized prior to pregnancy. Several risk factors of GDM depend on environmental factors that are thought to regulate the genome through epigenetic mechanisms. Thus, epigenetic regulation could be involved in the development of GDM. In addition, the adverse intrauterine environment in patients with GDM could also have a negative impact on the establishment of the epigenomes of the offspring.

15.
Oncogene ; 28(11): 1477-84, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19169274

ABSTRACT

Histone deacetylases (HDACs) play a key role in the regulation of gene expression and chromatin structure, and drugs targeting these enzymes might have an important impact in the treatment of human cancer. Herein, we report the characterization of (1H)-pyrroles as a new subfamily of HDAC inhibitors obtained by computational modeling of class-I human HDACs. From a functional standpoint, (1H)-pyrroles are powerful inductors of acetylation of histones H3 and H4, and restore the expression of growth-inhibitory genes. From a cellular view, these compounds cause a marked decrease in the viability of cancer cells in vitro and in vivo, associated with a cell-cycle arrest at G2/M and an inhibition of angiogenesis. Thus, (1H)-pyrroles emerge as a novel group of HDAC inhibitors with promising antitumoral features.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Pyrroles/pharmacology , Animals , Cell Line, Tumor , Computer Simulation , Dose-Response Relationship, Drug , Humans , Hydroxamic Acids/pharmacology , Mice , Models, Molecular , Structure-Activity Relationship , Vorinostat , Xenograft Model Antitumor Assays
16.
Oncogene ; 28(6): 781-91, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19060927

ABSTRACT

Sirtuin 1 (Sirt1) and Sirtuin 2 (Sirt2) belong to the family of NAD+ (nicotinamide adenine dinucleotide-positive)-dependent class III histone deacetylases and are involved in regulating lifespan. As cancer is a disease of ageing, targeting Sirtuins is emerging as a promising antitumour strategy. Here we present Salermide (N-{3-[(2-hydroxy-naphthalen-1-ylmethylene)-amino]-phenyl}-2-phenyl-propionamide), a reverse amide with a strong in vitro inhibitory effect on Sirt1 and Sirt2. Salermide was well tolerated by mice at concentrations up to 100 muM and prompted tumour-specific cell death in a wide range of human cancer cell lines. The antitumour activity of Salermide was primarily because of a massive induction of apoptosis. This was independent of global tubulin and K16H4 acetylation, which ruled out a putative Sirt2-mediated apoptotic pathway and suggested an in vivo mechanism of action through Sirt1. Consistently with this, RNA interference-mediated knockdown of Sirt1, but not Sirt2, induced apoptosis in cancer cells. Although p53 has been reported to be a target of Sirt1, genetic p53 knockdowns showed that the Sirt1-dependent proapoptotic effect of Salermide is p53-independent. We were finally able to ascribe the apoptotic effect of Salermide to the reactivation of proapoptotic genes epigenetically repressed exclusively in cancer cells by Sirt1. Taken together, our results underline Salermide's promise as an anticancer drug and provide evidence for the molecular mechanism through which Sirt1 is involved in human tumorigenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Naphthols/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Phenylpropionates/pharmacology , Sirtuins/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Genes, p53 , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Naphthols/chemistry , Phenylpropionates/chemistry , Sirtuin 1 , Sirtuin 2 , Sirtuins/metabolism , Tumor Suppressor Protein p53/metabolism
17.
Oncogene ; 27(54): 6817-33, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18762811

ABSTRACT

Hypomethylation of repeated elements in the genome is a common feature of human cancer, however, the direct consequences of this epigenetic defect for cancer biology are still largely unknown. Telomeres are specialized chromatin structures at the ends of eukaryotic chromosomes formed by tandem repeats of G-rich sequences and associated proteins, which have an essential role in chromosome end protection and genomic stability. Telomeric DNA repeats cannot be methylated, however, the adjacent subtelomeric DNA is heavily methylated in humans. Here, we show that the methylation status of subtelomeric DNA repeats negatively correlates with telomere length and telomere recombination in a large panel of human cancer cell lines. These findings suggest that tumor telomere length and integrity can be influenced by epigenetic factors. Finally, we show that treatment of human cancer cell lines with demethylating drugs results in hypomethylation of subtelomeric repeats and increased telomere recombination, which in turn may facilitate telomere elongation. All together, these findings suggest that tumor telomere length and integrity can be influenced by the epigenetic status of cancer cells.


Subject(s)
Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Telomere/genetics , Acetylation , Cell Line, Tumor , DNA Methylation/genetics , DNA, Neoplasm/genetics , Gene Amplification , Genome, Human , Histones/genetics , Humans , Recombination, Genetic , Repetitive Sequences, Nucleic Acid/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere/chemistry , Telomere/ultrastructure
18.
Rev. Soc. Bras. Cir. Craniomaxilofac ; 11(3,supl): 18-18, jun. 2008.
Article in Portuguese | LILACS | ID: lil-523549

ABSTRACT

Introdução: A lipomatose congênita infiltrativa caracteriza-se pela apresentação de tumores benignos não-encapsulados, que infiltram musculatura e partes moles adjacentes. O diagnóstico está baseado em aspectos clínicos, radiológicos e histológicos. Persistem controvérsias na literatura a respeito da história natural e tratamento desta condição, que apresenta elevadas taxas de recidiva. Relato do caso: Apresentamos o caso de um paciente masculino, 2 anos, submetido a ressecção parcial do tumor, realizada através de um acesso nasogeniano estendido, objetivando melhora estética. No acompanhamento posterior, havia sinais clínicos de crescimento já a partir do 3o. mês pós-operatório.


Subject(s)
Humans , Lipomatosis/surgery , Neoplasms/surgery , Neoplasms/diagnosis
19.
Oncogene ; 27(36): 4969-72, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18427547

ABSTRACT

Spry2 has been characterized as a negative regulator of the extracellular-regulated kinase (ERK) pathway. In this study we analysed whether epigenetic alterations of hSpry2 promoter occur in human lymphoid/hematopoietic malignancies. Our results revealed that hSpry2 promoter was hypermethylated in the HT cell line derived from a B-cell diffuse lymphoma, which correlated with decreased hSpry2 expression. We detected deregulation of the ERK pathway in these cells, but not in other blood cell lines expressing hSpry2. In addition, the ectopic overexpression of hSpry2 in HT cells drastically reduced the activation of ERK upon phorbol 12-myristate-13-acetate stimulation. Nude mice inoculated with HT mock cells developed tumors seven times larger than those from HT-hSpry2-transfected cells. We found hypermethylation of hSpry2 promoter in 37% (26 cases out of 71) of primary tumors from patients with B-cell diffuse lymphoma but none in normal B lymphocytes from 37 healthy individuals. Finally, we detected that hSpry2 promoter hypermethylation was associated with a significant decrease in the 5-year survival rate. These data suggest that hSpry2 could be important in lymphoid malignancies.


Subject(s)
Epigenesis, Genetic , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/physiology , Lymphoma, B-Cell/genetics , Protein Kinase Inhibitors/pharmacology , Animals , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lymphoma, B-Cell/enzymology , Membrane Proteins , Mice , Mice, Nude , Tetradecanoylphorbol Acetate/pharmacology
20.
Oncogene ; 27(25): 3556-66, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18223687

ABSTRACT

Methyl-cytosine-phosphate-guanine (CpG)-binding domain (MBD) proteins are bound to hypermethylated promoter CpG islands of tumor suppressor genes in human cancer cells, although a direct causal relationship at the genome-wide level between MBD presence and gene silencing remains to be demonstrated. To this end, we have inhibited the expression of MBD proteins in HeLa cells by short hairpin RNAs; and studied the functional consequences of MBD depletion using microarray-based expression analysis in conjunction with extensive bisulfite genomic sequencing and chromatin immunoprecipitation. The removal of MBDs results in a release of gene silencing associated with a loss of MBD occupancy in 5'-CpG islands without any change in the DNA methylation pattern. Our results unveil new targets for epigenetic inactivation mediated by MBDs in transformed cells, such as the cell adhesion protein gamma-parvin and the fibroblast growth factor 19, where we also demonstrate their bona fide tumor suppressor features. Our data support a fundamental role for MBD proteins in the direct maintenance of transcriptional repression of tumor suppressors and identify new candidate genes for epigenetic disruption in cancer cells.


Subject(s)
CpG Islands , Epigenesis, Genetic , Gene Silencing , Genes, Tumor Suppressor , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , Fibroblast Growth Factors/metabolism , HeLa Cells , Humans , Models, Biological , Protein Binding , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...