Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Rep ; 40(7): 111201, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977482

ABSTRACT

Stimulatory type 1 conventional dendritic cells (cDC1s) engage in productive interactions with CD8+ effectors along tumor-stroma boundaries. The paradoxical accumulation of "poised" cDC1s within stromal sheets is unlikely to simply reflect passive exclusion from tumor cores. Drawing parallels with embryonic morphogenesis, we hypothesized that invasive margin stromal remodeling generates developmentally conserved cell fate cues that regulate cDC1 behavior. We find that, in human T cell-inflamed tumors, CD8+ T cells penetrate tumor nests, whereas cDC1s are confined within adjacent stroma that recurrently displays site-specific proteolysis of the matrix proteoglycan versican (VCAN), an essential organ-sculpting modification in development. VCAN is necessary, and its proteolytic fragment (matrikine) versikine is sufficient for cDC1 accumulation. Versikine does not influence tumor-seeding pre-DC differentiation; rather, it orchestrates a distinctive cDC1 activation program conferring exquisite sensitivity to DNA sensing, supported by atypical innate lymphoid cells. Thus, peritumoral stroma mimicking embryonic provisional matrix remodeling regulates cDC1 abundance and activity to elicit T cell-inflamed tumor microenvironments.


Subject(s)
Neoplasms , Tumor Microenvironment , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Humans , Immunity, Innate , Lymphocytes/metabolism , Neoplasms/pathology , Versicans/metabolism
2.
Am J Physiol Cell Physiol ; 323(3): C678-C693, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35876288

ABSTRACT

Cancer immunoediting progresses through elimination, equilibrium, and escape. Each of these phases is characterized by breaching, remodeling, and rebuilding tissue planes and structural barriers that engage extracellular matrix (ECM) components, in particular matrix proteoglycans. Some of the signals emanating from matrix proteoglycan remodeling are readily co-opted by the growing tumor to sustain an environment of tumor-promoting and immune-suppressive inflammation. Yet other matrix-derived cues can be viewed as part of a homeostatic response by the host, aiming to eliminate the tumor and restore tissue integrity. These latter signals may be harnessed for therapeutic purposes to tip the polarity of the tumor immune milieu toward anticancer immunity. In this review, we attempt to showcase the importance and complexity of matrix proteoglycan signaling in both cancer-restraining and cancer-promoting inflammation. We propose that the era of matrix diagnostics and therapeutics for cancer is fast approaching the clinic.


Subject(s)
Neoplasms , Proteoglycans , Extracellular Matrix/pathology , Humans , Inflammation , Neoplasms/pathology , Signal Transduction
4.
Nature ; 588(7838): 491-497, 2020 12.
Article in English | MEDLINE | ID: mdl-33149299

ABSTRACT

Interferon-induced transmembrane protein 3 (IFITM3) has previously been identified as an endosomal protein that blocks viral infection1-3. Here we studied clinical cohorts of patients with B cell leukaemia and lymphoma, and identified IFITM3 as a strong predictor of poor outcome. In normal resting B cells, IFITM3 was minimally expressed and mainly localized in endosomes. However, engagement of the B cell receptor (BCR) induced both expression of IFITM3 and phosphorylation of this protein at Tyr20, which resulted in the accumulation of IFITM3 at the cell surface. In B cell leukaemia, oncogenic kinases phosphorylate IFITM3 at Tyr20, which causes constitutive localization of this protein at the plasma membrane. In a mouse model, Ifitm3-/- naive B cells developed in normal numbers; however, the formation of germinal centres and the production of antigen-specific antibodies were compromised. Oncogenes that induce the development of leukaemia and lymphoma did not transform Ifitm3-/- B cells. Conversely, the phosphomimetic IFITM3(Y20E) mutant induced oncogenic PI3K signalling and initiated the transformation of premalignant B cells. Mechanistic experiments revealed that IFITM3 functions as a PIP3 scaffold and central amplifier of PI3K signalling. The amplification of PI3K signals depends on IFITM3 using two lysine residues (Lys83 and Lys104) in its conserved intracellular loop as a scaffold for the accumulation of PIP3. In Ifitm3-/- B cells, lipid rafts were depleted of PIP3, which resulted in the defective expression of over 60 lipid-raft-associated surface receptors, and impaired BCR signalling and cellular adhesion. We conclude that the phosphorylation of IFITM3 that occurs after B cells encounter antigen induces a dynamic switch from antiviral effector functions in endosomes to a PI3K amplification loop at the cell surface. IFITM3-dependent amplification of PI3K signalling, which in part acts downstream of the BCR, is critical for the rapid expansion of B cells with high affinity to antigen. In addition, multiple oncogenes depend on IFITM3 to assemble PIP3-dependent signalling complexes and amplify PI3K signalling for malignant transformation.


Subject(s)
B-Lymphocytes/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Animals , Antigens, CD19/metabolism , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Transformation, Neoplastic , Female , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/pathology , Humans , Integrins/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Models, Molecular , Phosphorylation , Receptors, Antigen, B-Cell/metabolism
5.
Nature ; 583(7818): 845-851, 2020 07.
Article in English | MEDLINE | ID: mdl-32699415

ABSTRACT

Malignant transformation of cells typically involves several genetic lesions, whose combined activity gives rise to cancer1. Here we analyse 1,148 patient-derived B-cell leukaemia (B-ALL) samples, and find that individual mutations do not promote leukaemogenesis unless they converge on one single oncogenic pathway that is characteristic of the differentiation stage of transformed B cells. Mutations that are not aligned with this central oncogenic driver activate divergent pathways and subvert transformation. Oncogenic lesions in B-ALL frequently mimic signalling through cytokine receptors at the pro-B-cell stage (via activation of the signal-transduction protein STAT5)2-4 or pre-B-cell receptors in more mature cells (via activation of the protein kinase ERK)5-8. STAT5- and ERK-activating lesions are found frequently, but occur together in only around 3% of cases (P = 2.2 × 10-16). Single-cell mutation and phospho-protein analyses reveal the segregation of oncogenic STAT5 and ERK activation to competing clones. STAT5 and ERK engage opposing biochemical and transcriptional programs that are orchestrated by the transcription factors MYC and BCL6, respectively. Genetic reactivation of the divergent (suppressed) pathway comes at the expense of the principal oncogenic driver and reverses transformation. Conversely, deletion of divergent pathway components accelerates leukaemogenesis. Thus, persistence of divergent signalling pathways represents a powerful barrier to transformation, while convergence on one principal driver defines a central event in leukaemia initiation. Pharmacological reactivation of suppressed divergent circuits synergizes strongly with inhibition of the principal oncogenic driver. Hence, reactivation of divergent pathways can be leveraged as a previously unrecognized strategy to enhance treatment responses.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Transformation, Neoplastic , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Signal Transduction , Animals , B-Lymphocytes/pathology , Cell Line, Tumor , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Proto-Oncogene Proteins c-myc/metabolism , STAT5 Transcription Factor/metabolism
6.
Leukemia ; 34(5): 1266-1277, 2020 05.
Article in English | MEDLINE | ID: mdl-31780813

ABSTRACT

The histone demethylase lysine-specific demethylase 1 (LSD1 or KDM1A) has emerged as a candidate therapeutic target in acute myeloid leukaemia (AML); tranylcypromine-derivative inhibitors induce loss of clonogenic activity and promote differentiation, in particular in the MLL-translocated molecular subtype of AML. In AML, the use of drugs in combination often delivers superior clinical activity. To identify genes and cellular pathways that collaborate with LSD1 to maintain the leukaemic phenotype, and which could be targeted by combination therapies, we performed a genome-wide CRISPR-Cas9 dropout screen. We identified multiple components of the amino acid sensing arm of mTORC1 signalling-RRAGA, MLST8, WDR24 and LAMTOR2-as cellular sensitizers to LSD1 inhibition. Knockdown of mTORC1 components, or mTORC1 pharmacologic inhibition, in combination with LSD1 inhibition enhanced differentiation in both cell line and primary cell settings, in vitro and in vivo, and substantially reduced the frequency of clonogenic primary human AML cells in a modelled minimal residual disease setting. Synergistic upregulation of a set of transcription factor genes associated with terminal monocytic lineage differentiation was observed. Thus, dual mTORC1 and LSD1 inhibition represents a candidate combination approach for enhanced differentiation in MLL-translocated AML which could be evaluated in early phase clinical trials.


Subject(s)
Everolimus/pharmacology , Histone Demethylases/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/drug therapy , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Myeloid-Lymphoid Leukemia Protein/genetics , Translocation, Genetic , Tranylcypromine/pharmacology , Animals , Antidepressive Agents/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Drug Therapy, Combination , Female , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Genes Dev ; 33(17-18): 1265-1279, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31395741

ABSTRACT

Chromosomal rearrangements of the mixed lineage leukemia (MLL) gene occur in ∼10% of B-cell acute lymphoblastic leukemia (B-ALL) and define a group of patients with dismal outcomes. Immunohistochemical staining of bone marrow biopsies from most of these patients revealed aberrant expression of BCL6, a transcription factor that promotes oncogenic B-cell transformation and drug resistance in B-ALL. Our genetic and ChIP-seq (chromatin immunoprecipitation [ChIP] combined with high-throughput sequencing) analyses showed that MLL-AF4 and MLL-ENL fusions directly bound to the BCL6 promoter and up-regulated BCL6 expression. While oncogenic MLL fusions strongly induced aberrant BCL6 expression in B-ALL cells, germline MLL was required to up-regulate Bcl6 in response to physiological stimuli during normal B-cell development. Inducible expression of Bcl6 increased MLL mRNA levels, which was reversed by genetic deletion and pharmacological inhibition of Bcl6, suggesting a positive feedback loop between MLL and BCL6. Highlighting the central role of BCL6 in MLL-rearranged B-ALL, conditional deletion and pharmacological inhibition of BCL6 compromised leukemogenesis in transplant recipient mice and restored sensitivity to vincristine chemotherapy in MLL-rearranged B-ALL patient samples. Oncogenic MLL fusions strongly induced transcriptional activation of the proapoptotic BH3-only molecule BIM, while BCL6 was required to curb MLL-induced expression of BIM. Notably, peptide (RI-BPI) and small molecule (FX1) BCL6 inhibitors derepressed BIM and synergized with the BH3-mimetic ABT-199 in eradicating MLL-rearranged B-ALL cells. These findings uncover MLL-dependent transcriptional activation of BCL6 as a previously unrecognized requirement of malignant transformation by oncogenic MLL fusions and identified BCL6 as a novel target for the treatment of MLL-rearranged B-ALL.


Subject(s)
Gene Expression Regulation, Leukemic , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Survival/genetics , Cells, Cultured , Gene Deletion , Gene Targeting , Humans , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prognosis , Promoter Regions, Genetic/genetics
8.
Mol Carcinog ; 58(7): 1194-1207, 2019 07.
Article in English | MEDLINE | ID: mdl-30854739

ABSTRACT

Green tea polyphenols (GTPs) and their major constituent, epigallocatechin-3-gallate (EGCG), have been reported to demonstrate many interesting biological activities, including anticancer properties. Recent studies on prostate cancer provide strong evidence that epigenetic mechanisms are major players in the regulation of matrix metalloproteinases (MMPs) and their binding partner tissue inhibitor of MMPs (TIMPs) involved in prostate cancer progression. Here we demonstrate that GTP/EGCG mediate epigenetic reactivation of TIMP-3 that plays a key role in suppressing invasiveness and cancer progression. Treatment of human prostate cancer DUPRO and LNCaP cells with 10 µg/mL GTP and 20 µM EGCG induced TIMP-3 mRNA and protein expression. This transcriptional activation of TIMP-3 was associated with the decrease in the expression of both enhancers of zeste homolog 2 (EZH2) and its catalytic product trimethylation of histone H3 at lysine 27 (H3K27me3) repressive marks at the TIMP-3 promoter with an accompanying increase in histone H3K9/18 acetylation. In addition, GTP/EGCG treatment significantly reduced class I histone deacetylase (HDAC) activity/expression and EZH2 and H3K27me3 levels in prostate cancer cells. EGCG/GTP exposure also reduced MMP-2/MMP-9 gelatinolytic activity and abrogated invasion and migration capabilities in these cells. Silencing of EZH2 and class I HDACs strikingly increased the expression of TIMP-3 independent of DNA methylation. Furthermore, clinical trials performed on patients undergoing prostatectomy consuming 800 mg EGCG (Polyphenon E) up to 6 weeks and grade-matched controls demonstrate an increase in plasma TIMP-3 levels. A marked reduction in class I HDACs activity/expression and EZH2 and H3K27me3 levels were noted in GTP-supplemented prostate tissue. Our findings highlight that TIMP-3 induction, as a key epigenetic event modulated by green tea in restoring the MMP:TIMP balance suppresses prostate cancer progression.


Subject(s)
Antineoplastic Agents/therapeutic use , Catechin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Tea/chemistry , Tissue Inhibitor of Metalloproteinase-3/metabolism , Acetylation/drug effects , Catechin/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , DNA Methylation/drug effects , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Histone Code/drug effects , Histone Code/physiology , Histone Deacetylase 1/metabolism , Histones/biosynthesis , Humans , Male , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness/pathology , Plant Preparations/therapeutic use , Polyphenols/therapeutic use , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-3/blood , Tissue Inhibitor of Metalloproteinase-3/genetics , Transcriptional Activation/drug effects
9.
J Med Chem ; 60(2): 668-680, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28068087

ABSTRACT

The BRPF (bromodomain and PHD finger-containing) family are scaffolding proteins important for the recruitment of histone acetyltransferases of the MYST family to chromatin. Evaluation of the BRPF family as a potential drug target is at an early stage although there is an emerging understanding of a role in acute myeloid leukemia (AML). We report the optimization of fragment hit 5b to 13-d as a biased, potent inhibitor of the BRD of the BRPFs with excellent selectivity over nonclass IV BRD proteins. Evaluation of 13-d in a panel of cancer cell lines showed a selective inhibition of proliferation of a subset of AML lines. Pharmacokinetic studies established that 13-d had properties compatible with oral dosing in mouse models of disease (Fpo 49%). We propose that NI-42 (13-d) is a new chemical probe for the BRPFs suitable for cellular and in vivo studies to explore the fundamental biology of these proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Nuclear Proteins/antagonists & inhibitors , Quinolones/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , DNA-Binding Proteins , Drug Screening Assays, Antitumor , Humans , Leukemia, Myeloid, Acute/drug therapy , Mice , Microsomes, Liver/metabolism , Protein Domains , Quinolones/chemical synthesis , Quinolones/chemistry , Quinolones/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics
10.
Cancer Cell ; 30(1): 5-7, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27411580

ABSTRACT

E proteins and their functional antagonists, the ID proteins, have significant roles in normal hematopoiesis. In this issue of Cancer Cell, Ghisi et al. show that high ID2 levels antagonize self-renewal and promote differentiation of leukemic stem cells in the MLL-translocated molecular subtype of acute myeloid leukemia.


Subject(s)
Cell Differentiation , Leukemia, Myeloid, Acute/genetics , Hematopoietic Stem Cells/metabolism , Humans , Transcription Factors , Translocation, Genetic
11.
Data Brief ; 6: 461-5, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26977427

ABSTRACT

In this data article we have provided evidence for the negative influence of divalent cations on (-)-epigallocatechin-3-gallate (EGCG)-mediated inhibition of matrix metalloproteinase-2 (MMP-2) activity in cell-free experiments. Chelating agents, such as EDTA and sodium citrate alone, did not affect MMP-2 activity. While EDTA enhanced, excess of divalent cations interfered with EGCG-mediated inhibition of MMP-2.

12.
Genom Data ; 5: 210-2, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26484257

ABSTRACT

(-)-Epigallocatechin-3-gallate (EGCG) is one of the most potent and the most studied green tea catechin. Reports on mechanisms of EGCG action and its cellular targets are plenty. Compelling evidences in the literature in favor of ER being one of its targets suggest that EGCG may have a significant impact on estrogen regulated gene expression. Despite the possible implications on breast cancer chemoprevention or therapy, this aspect of EGCG action has not been adequately investigated. In order to address this issue, we have obtained gene expression profiles of MCF-7 breast cancer cells treated with ethanol (vehicle control) and those treated with estrogen, EGCG or both, using microarrays. Here, we have presented in detail the design and execution of the microarray experiment, quality control checks and analysis of microarray data. The utility and importance of the data generated in this work have been discussed in the context of the background literature. Our data is available in the Gene Expression Omnibus (GEO) database with the identifier GSE56245.

13.
Mol Carcinog ; 54(6): 485-99, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24481780

ABSTRACT

Aberrant epigenetic silencing of the tissue inhibitor of matrix metalloproteinase-3 (TIMP-3) gene that negatively regulates matrix metalloproteinases (MMPs) activity has been implicated in the pathogenesis and metastasis of breast cancer. In the present study, we demonstrate that green tea polyphenols (GTP) and its major constituent, epigallocatechin-3-gallate (EGCG) mediate epigenetic induction of TIMP-3 levels and play a key role in suppressing invasiveness and gelatinolytic activity of MMP-2 and MMP-9 in breast cancer cells. Treatment of MCF-7 and MDA-MB-231 breast cancer cells with 20 µM EGCG and 10 µg/mL GTP for 72 h significantly induces TIMP-3 mRNA and protein levels. Interestingly, investigations into the molecular mechanism revealed that TIMP-3 repression in breast cancer cells is mediated by epigenetic silencing mechanism(s) involving increased activity of the enhancer of zeste homolog 2 (EZH2) and class I histone deacetylases (HDACs), independent of promoter DNA hypermethylation. Treatment of breast cancer cells with GTP and EGCG significantly reduced EZH2 and class I HDAC protein levels. Furthermore, transcriptional activation of TIMP-3 was found to be associated with decreased EZH2 localization and H3K27 trimethylation enrichment at the TIMP-3 promoter with a concomitant increase in histone H3K9/18 acetylation. Our findings highlight TIMP-3 induction as a key epigenetic event modulated by GTPs in restoring the MMP:TIMP balance to delay breast cancer progression and invasion.


Subject(s)
Breast Neoplasms/genetics , Catechin/analogs & derivatives , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Polyphenols/pharmacology , Tissue Inhibitor of Metalloproteinase-3/genetics , Acetylation/drug effects , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Catechin/chemistry , Catechin/pharmacology , Cell Line, Tumor , DNA Methylation/drug effects , Female , Gene Silencing/drug effects , Humans , Polyphenols/chemistry , Promoter Regions, Genetic/drug effects , Tea/chemistry
14.
Indian J Exp Biol ; 52(4): 312-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24772933

ABSTRACT

Investigations using in vitro and in vivo models of breast carcinogenesis have demonstrated anti-neoplastic activity of the green tea polyphenol, epigallocatechin gallate (EGCG). Although a number of molecular targets of EGCG have been identified, its impact on the expression of estrogen target genes is not completely understood. Here, we examined the mRNA expression levels of two estrogen target genes, namely Trefoil Factor 1 (pS2) and Progesterone Receptor (PR) in MCF-7 cells treated with EGCG. We observed that treatment with 40 microM EGCG, which caused only 20% decrease in cell viability, resulted in increased steady state expression levels of pS2 and PR mRNA. This suggests that EGCG may exert its biological activities, at least in part, by influencing the expression of estrogen target genes.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/genetics , Catechin/analogs & derivatives , Receptors, Progesterone/genetics , Tumor Suppressor Proteins/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catechin/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Progesterone/metabolism , Trefoil Factor-1 , Tumor Suppressor Proteins/metabolism
15.
Mol Cancer Res ; 12(5): 639-53, 2014 May.
Article in English | MEDLINE | ID: mdl-24526064

ABSTRACT

Seminal discoveries have established that epigenetic modifications are important for driving tumor progression. Polycomb group (PcG) proteins are highly conserved epigenetic effectors that maintain, by posttranslational modification of histones, the silenced state of genes involved in critical biologic processes, including cellular development, stem cell plasticity, and tumor progression. PcG proteins are found in two multimeric protein complexes called Polycomb repressive complexes: PRC1 and PRC2. Enhancer of zeste homolog 2 (EZH2), catalytic core subunit of PRC2, epigenetically silences several tumor-suppressor genes by catalyzing the trimethylation of histone H3 at lysine 27, which serves as a docking site for DNA methyltransferases and histone deacetylases. Evidence suggests that overexpression of EZH2 is strongly associated with cancer progression and poor outcome in disparate cancers, including hematologic and epithelial malignancies. The regulatory circuit and molecular cues causing EZH2 deregulation vary in different cancer types. Therefore, this review provides a comprehensive overview on the oncogenic role of EZH2 during tumorigenesis and highlights the multifaceted role of EZH2, as either a transcriptional activator or repressor depending on the cellular context. Additional insight is provided on the recent understanding of the causes and consequences of EZH2 overexpression in specific cancer types. Finally, evidence is discussed on how EZH2 has emerged as a promising target in anticancer therapy and the prospects for targeting EZH2 without affecting global methylation status. Thus, a better understanding of the complex epigenetic regulatory network controlling EZH2 expression and target genes facilitates the design of novel therapeutic interventions.


Subject(s)
Polycomb Repressive Complex 2/genetics , Animals , Cell Transformation, Neoplastic/genetics , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Humans , Polycomb Repressive Complex 2/metabolism
16.
AAPS J ; 16(1): 151-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24307610

ABSTRACT

In recent years, "nutri-epigenetics," which focuses on the influence of dietary agents on epigenetic mechanism(s), has emerged as an exciting novel area in epigenetics research. Targeting of aberrant epigenetic modifications has gained considerable attention in cancer chemoprevention research because, unlike genetic changes, epigenetic alterations are reversible and occur during early carcinogenesis. Aberrant epigenetic mechanisms, such as promoter DNA methylation, histone modifications, and miRNA-mediated post-transcriptional alterations, can silence critical tumor suppressor genes, such as transcription factors, cell cycle regulators, nuclear receptors, signal transducers, and apoptosis-inducing and DNA repair gene products, and ultimately contribute to carcinogenesis. In an effort to identify and develop anticancer agents which cause minimal harm to normal cells while effectively killing cancer cells, a number of naturally occurring phytochemicals in food and medicinal plants have been investigated. This review highlights the potential role of plant-derived phytochemicals in targeting epigenetic alterations that occur during carcinogenesis, by modulating the activity or expression of DNA methyltransferases, histone modifying enzymes, and miRNAs. We present in detail the epigenetic mode of action of various phytochemicals and discuss their potential as safe and clinically useful chemopreventive strategies.


Subject(s)
Epigenesis, Genetic/drug effects , Neoplasms/prevention & control , Phytochemicals/therapeutic use , Phytotherapy , Animals , Chemoprevention , Diet , Diet Therapy , Humans , Phytochemicals/pharmacology
17.
Epigenetics ; 8(5): 464-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23644490

ABSTRACT

Overexpression of EZH2 and other PRC2 subunits, such as SUZ12, is associated with tumor progression and poor prognosis in several human malignancies. Nevertheless, the underlying mechanisms driving aberrant EZH2 expression are poorly understood. This review provides molecular insights into the essential role of EZH2 in breast and prostate tumorigenesis. We addressed the current understanding on the oncogenic role of EZH2, with an emphasis on: (1) the less known PRC2-independent role of EZH2 in gene activation, in addition to its canonical role in transcriptional silencing as a histone methyltransferase catalyzing the trimethylation of histone H3 at lysine 27; (2) causes and consequences of its deregulation in tumor cells and; (3) collaboration of EZH2 with other epigenetic and hormone receptor-mediated oncogenic signaling pathways. We also summarize how EZH2 has emerged as a promising therapeutic target in hormone-refractory cancers and the prospects for integrating EZH2 blockade with available pharmacological inhibitors.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Epigenesis, Genetic , Polycomb Repressive Complex 2/metabolism , Prostatic Neoplasms/genetics , Female , Histones/metabolism , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...