Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
1.
Adv Cancer Res ; 130: 55-111, 2016.
Article in English | MEDLINE | ID: mdl-27037751

ABSTRACT

Epigenetic silencing and inappropriate activation of gene expression are frequent events during the initiation and progression of cancer. These events involve a complex interplay between the hypermethylation of CpG dinucleotides within gene promoter and enhancer regions, the recruitment of transcriptional corepressors and the deacetylation and/or methylation of histone tails. These epigenetic regulators act in concert to block transcription or interfere with the maintenance of chromatin boundary regions. However, DNA/histone methylation and histone acetylation states are reversible, enzyme-mediated processes and as such, have emerged as promising targets for cancer therapy. This review will focus on the potential benefits and synergistic/additive effects of combining DNA-demethylating agents and histone deacetylase inhibitors or lysine-specific demethylase inhibitors together in epigenetic therapy for solid tumors and will highlight what is known regarding the mechanisms of action that contribute to the antitumor response.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA Methylation/genetics , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/therapeutic use , Histones/metabolism , Neoplasms/drug therapy , Drug Synergism , Epigenesis, Genetic , Humans , Promoter Regions, Genetic , RNA/genetics , RNA/metabolism , Transcription, Genetic
2.
Oncogene ; 33(17): 2157-68, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-23708667

ABSTRACT

Many tumor suppressor genes (TSGs) are silenced through synergistic layers of epigenetic regulation including abnormal DNA hypermethylation of promoter CpG islands, repressive chromatin modifications and enhanced nucleosome deposition over transcription start sites. The protein complexes responsible for silencing of many of such TSGs remain to be identified. Our previous work demonstrated that multiple silenced TSGs in colorectal cancer cells can be partially reactivated by DNA demethylation in cells disrupted for the DNA methyltransferases 1 and 3B (DNMT1 and 3B) or by DNMT inhibitors (DNMTi). Herein, we used proteomic and functional genetic approaches to identify additional proteins that cooperate with DNMTs in silencing these key silenced TSGs in colon cancer cells. We discovered that DNMTs and the core components of the NuRD (Mi-2/nucleosome remodeling and deacetylase) nucleosome remodeling complex, chromo domain helicase DNA-binding protein 4 (CHD4) and histone deacetylase 1 (HDAC1) occupy the promoters of several of these hypermethylated TSGs and physically and functionally interact to maintain their silencing. Consistent with this, we find an inverse relationship between expression of HDAC1 and 2 and these TSGs in a large panel of primary colorectal tumors. We demonstrate that DNMTs and NuRD cooperate to maintain the silencing of several negative regulators of the WNT and other signaling pathways. We find that depletion of CHD4 is synergistic with DNMT inhibition in reducing the viability of colon cancer cells in correlation with reactivation of TSGs, suggesting that their combined inhibition may be beneficial for the treatment of colon cancer. Since CHD4 has ATPase activity, our data identify CHD4 as a potentially novel drug target in cancer.


Subject(s)
Autoantigens/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Apoptosis , Autoantigens/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Colorectal Neoplasms , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Decitabine , Epigenesis, Genetic , Gene Expression , Gene Knockdown Techniques , Genes, Tumor Suppressor , HCT116 Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Promoter Regions, Genetic , Protein Binding , Wnt Proteins/metabolism
3.
Oncogene ; 30(23): 2659-69, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21297660

ABSTRACT

Adenomatous polyposis coli (APC) gene mutations have been implicated in familial and sporadic gastrointestinal (GI) cancers. APC mutations are associated with autosomal dominant inheritance of disease in humans. Similarly, mice that contain a single mutant APC gene encoding a protein truncated at residue 716 (Apc(Δ716)) develop multiple polyps throughout the GI tract as early as 4 weeks after birth. Inactivation of another tumor suppressor gene, Hypermethylated in Cancer 1 (HIC1), often occurs in human colon cancers, among others, via CpG island hypermethylation. Homozygous deletion of Hic1 in mice results in major developmental defects and embryonic lethality. Hic1 heterozygotes have previously been shown to develop tumors of a variety of tissue types. We now report that loss of a single Hic1 allele can promote crypt hyperplasia and neoplasia of the GI tract, and Hic1(+/-), Apc(+/Δ716) double heterozygotes (DH) develop increased numbers of polyps throughout the GI tract at 60 days. Hic1 expression is absent in polyps from DH mice, with concomitant increased expression of two transcriptional repression targets of Hic1, Sirt1 and Sox9. Together, our data suggest that loss of a gene frequently silenced via epigenetic mechanisms, Hic1, can cooperate with loss of a gene mutated in GI cancer, Apc, to promote tumorigenesis in an in vivo model of multiple intestinal neoplasia.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Intestine, Small/metabolism , Kruppel-Like Transcription Factors/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cells, Cultured , CpG Islands/genetics , DNA Methylation , Embryo, Mammalian/cytology , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Heterozygote , Humans , Hyperplasia , Immunohistochemistry , Intestine, Small/pathology , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
4.
Oncogene ; 29(44): 5923-34, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20697356

ABSTRACT

The Wnt signaling pathway is capable of self-regulation through positive and negative feedback mechanisms. For example, the oncoprotein c-Myc, which is upregulated by Wnt signaling activity, participates in a positive feedback loop of canonical Wnt signaling through repression of Wnt antagonists DKK1 and SFRP1. In this study, we investigated the mechanism of Wnt inhibitory factor-1 (WIF-1) silencing. Mapping of CpG island methylation of the WIF-1 promoter reveals regional methylation (-295 to -95 bp from the transcription start site) that correlates with transcriptional silencing. We identified Miz-1 as a direct activator of WIF-1 transcriptional activity, which is found at WIF-1 promoter. In addition, we show that c-Myc contributes to WIF-1 transcriptional repression in a Miz-1-dependent manner. Although the transient repression mediated by Miz-1/c-Myc is independent of de novo methylation, the stable repression by this complex is associated with CpG island methylation of the critical -295 to -95-bp region of the WIF-1 promoter. Importantly, Miz-1 and c-Myc are found at WIF-1 promoter in WIF-1 non-expressing cell lines DLD-1 and 209myc. Transient knockdown or somatic knockout of c-Myc in DLD-1 failed to restore WIF-1 expression suggesting that c-Myc is involved in initiating rather than maintaining WIF-1 epigenetic silencing. In a genome-wide screen, DNAJA4, TGFß-induced and TRIM59 were repressed by c-Myc overexpression and DNA promoter hypermethylation. Our data reveal novel insights into c-Myc-mediated DNA methylation-dependent transcriptional silencing, a mechanism that might contribute to the dysregulation of Wnt signaling in cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation/physiology , Kruppel-Like Transcription Factors/physiology , Proto-Oncogene Proteins c-myc/physiology , Repressor Proteins/genetics , Transcription, Genetic/physiology , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , Gene Silencing , Humans , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
5.
Oncogene ; 29(17): 2467-76, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20154726

ABSTRACT

The tumor suppressor gene hypermethylated in cancer 1 (HIC1), which encodes a transcriptional repressor, is epigenetically inactivated in various human cancers. In this study, we show that HIC1 is a direct transcriptional repressor of the gene encoding ephrin-A1, a cell surface ligand implicated in the pathogenesis of epithelial cancers. We also show that mouse embryos lacking both Hic1 alleles manifest developmental defects spatially associated with the misexpression of ephrin-A1, and that overexpression of ephrin-A1 is a feature of tumors arising in Hic1 heterozygous mice in which the remaining wild-type allele is epigenetically silenced. In breast cancer, we find that ephrin-A1 expression is common in vivo, but that in cell culture, expression of the EphA receptors is predominant. Restoration of HIC1 function in breast cancer cells leads to a reduction in tumor growth in vivo, an effect that can be partially rescued by co-overexpression of ephrin-A1. Interestingly, overexpression of ephrin-A1 in vitro triggers downregulation of EphA2 and EphA4 levels, resulting in an expression pattern similar to that seen in vivo. We conclude that Hic1 spatially restricts ephrin-A1 expression in development, and that upregulated expression of ephrin-A1 resulting from epigenetic silencing of HIC1 in cancer cells may be an important mechanism in epithelial malignancy.


Subject(s)
Breast Neoplasms/prevention & control , Ephrin-A1/genetics , Kruppel-Like Transcription Factors/physiology , Repressor Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Down-Regulation , Ephrin-A1/antagonists & inhibitors , Female , Humans , Mice
6.
Article in English | MEDLINE | ID: mdl-21447817

ABSTRACT

It is widely accepted that cancer results from an array of epigenetic and genetic alterations, particularly aberrant epigenetic patterns that are a hallmark of every cancer type studied. Another well-known feature of cancer cells is the array of abnormalities in their nuclear structure. Although it is known that nuclear structure has an important role in the regulation of gene expression, we know little about the direct relationship between nuclear structural alterations and aberrant epigenetic patterns in cancer. Here, we discuss some of the recent studies from our lab and others to understand the relationship between alterations of nuclear architecture and aberrant epigenetic patterns in cancer cells. Although the precise relationship remains elusive, we suggest that changes in nuclear structure and composition could alter long-range genomic interactions and cause global epigenetic changes during tumorigenesis. We emphasize the need for further studies to elucidate the direct relationship between nuclear structure alterations and aberrant epigenetic patterns in cancers.


Subject(s)
Cell Nucleus/chemistry , Cell Nucleus/genetics , Epigenesis, Genetic , Neoplasms/genetics , Neoplasms/pathology , Animals , Gene Expression Regulation, Neoplastic , Humans , Models, Genetic , Nuclear Proteins/metabolism
7.
Br J Cancer ; 98(6): 1147-56, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18283316

ABSTRACT

Although mutation of APC or CTNNB1 (beta-catenin) is rare in breast cancer, activation of Wnt signalling is nonetheless thought to play an important role in breast tumorigenesis, and epigenetic silencing of Wnt antagonist genes, including the secreted frizzled-related protein (SFRP) and Dickkopf (DKK) families, has been observed in various tumours. In breast cancer, frequent methylation and silencing of SFRP1 was recently documented; however, altered expression of other Wnt antagonist genes is largely unknown. In the present study, we found frequent methylation of SFRP family genes in breast cancer cell lines (SFRP1, 7 out of 11, 64%; SFRP2, 11 out of 11, 100%; SFRP5, 10 out of 11, 91%) and primary breast tumours (SFRP1, 31 out of 78, 40%; SFRP2, 60 out of 78, 77%; SFRP5, 55 out of 78, 71%). We also observed methylation of DKK1, although less frequently, in cell lines (3 out of 11, 27%) and primary tumours (15 out of 78, 19%). Breast cancer cell lines express various Wnt ligands, and overexpression of SFRPs inhibited cancer cell growth. In addition, overexpression of a beta-catenin mutant and depletion of SFRP1 using small interfering RNA synergistically upregulated transcriptional activity of T-cell factor/lymphocyte enhancer factor. Our results confirm the frequent methylation and silencing of Wnt antagonist genes in breast cancer, and suggest that their loss of function contributes to activation of Wnt signalling in breast carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Epigenesis, Genetic , Eye Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , DNA Methylation , Female , Gene Silencing , Genes, Tumor Suppressor , Humans
8.
Oncogene ; 25(18): 2666-73, 2006 Apr 27.
Article in English | MEDLINE | ID: mdl-16314833

ABSTRACT

Recently, it was reported that exogenous bone morphogenetic protein (BMP)-2 acted as an antiproliferative agent in a variety of cell lines, including normal and cancerous gastric cell lines, indicating that BMP-2 plays an important role during cell growth. However, despite the loss of BMP-2 expression in several cancers, the underlying mechanism remains unknown. Epigenetic silencing through DNA methylation is one of the key steps during carcinogenesis. In this study, we found, through analysis by the methylation-specific polymerase chain reaction technique, CpG island methylation of the BMP-2 promoter region in gastric and colon cancer cell lines. BMP-2 mRNA was found to be activated after 5-aza-2'-deoxycytidine treatment of the methylation-positive cells. Moreover, 24 of the 56 (42.9%) gastric cancer tissues exhibited promoter methylation. Immunohistochemical staining revealed that 18 of the 24 (75%) gastric cancer tissues without methylation signals exhibited BMP-2 expression, whereas among 20 cancer tissues with strong methylation signals only four (20%) expressed BMP-2 (P = 0.0003). These findings indicate that BMP-2 methylation is strongly associated with the loss of BMP-2 protein expression in the primary gastric carcinomas. BMP-2 methylation was more often observed in diffuse type (60.7%) than in intestinal type (25%) gastric carcinomas (P = 0.007). Thus, aberrant BMP-2 methylation and the resultant loss of BMP-2 expression may be related to gastric carcinogenesis, particularly in the diffuse type.


Subject(s)
Bone Morphogenetic Proteins/genetics , DNA Methylation , Epigenesis, Genetic , Gene Silencing , Stomach Neoplasms/genetics , Transforming Growth Factor beta/genetics , Aged , Base Sequence , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , CpG Islands/genetics , Diet , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Molecular Sequence Data , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
9.
Article in English | MEDLINE | ID: mdl-16869780

ABSTRACT

Although it is clear that genetic alterations are critical for the initiation and maintenance of human cancer, it is also becoming evident that epigenetic changes may be essential for the development of these diseases as well. The best studied of these latter processes is heritable transcriptional repression of genes associated with aberrant DNA hypermethylation of their promoters. Herein we review how very early occurrence of these gene silencing events may contribute to loss of key gene functions which result in disruption of cell regulatory pathways that may contribute to abnormal cell population expansion. These altered regulatory events may then provide a setting where mutations in the same disrupted pathways may be readily selected and serve to lock tumor progression into place. This hypothesis has potential impact on means to prevent and control cancer and for the use of epigenetic markers for cancer risk assessment and early diagnosis.


Subject(s)
Gene Silencing , Neoplasms/genetics , Animals , CpG Islands , DNA Methylation , Epigenesis, Genetic , Humans , Models, Genetic , Mutation , Neoplasms/etiology , Neoplasms/therapy
10.
Hum Mol Genet ; 10(26): 3001-7, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751682

ABSTRACT

Cancer cells have aberrant patterns of DNA methylation including hypermethylation of gene promoter CpG islands and global demethylation of the genome. Genes that cause familial cancer, as well as other genes, can be silenced by promoter hypermethylation in sporadic tumors, but the methylation of these genes in tumors from kindreds with inherited cancer syndromes has not been well characterized. Here, we examine CpG island methylation of 10 genes (hMLH1, BRCA1, APC, LKB1, CDH1, p16(INK4a), p14(ARF), MGMT, GSTP1 and RARbeta2) and 5-methylcytosine DNA content, in inherited (n = 342) and non-inherited (n = 215) breast and colorectal cancers. Our results show that singly retained alleles of germline mutated genes are never hypermethylated in inherited tumors. However, this epigenetic change is a frequent second "hit", associated with the wild-type copy of these genes in inherited tumors where both alleles are retained. Global hypomethylation was similar between sporadic and hereditary cases, but distinct differences existed in patterns of methylation at non-familial genes. This study demonstrates that hereditary cancers "mimic" the DNA methylation patterns present in the sporadic tumors.


Subject(s)
Breast Neoplasms/genetics , Colonic Neoplasms/genetics , DNA Methylation , Neoplastic Syndromes, Hereditary/genetics , Oncogenes , Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , CpG Islands , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Humans , Mutation , Neoplastic Syndromes, Hereditary/physiopathology , Promoter Regions, Genetic
11.
Oncogene ; 20(24): 3156-65, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11420732

ABSTRACT

Cancer is a process driven by the accumulation of abnormalities in gene function. While many of these changes are genetic, epigenetically mediated changes in gene expression are being increasingly appreciated. This latter process emphasizes the need to understand two key components of heritable, but reversible, modulation of gene promoter function that are closely tied to one another - formation of chromatin which modulates transcription and establishing patterns of DNA methylation. The link lies first in the recruitment to methylated cytosines of a family of methyl-CpG binding domain proteins (MBDs), which are direct transcriptional repressors and can complex with transcriptional corepressors including histone deacetylases (HDACs). Additionally, the proteins that catalyze DNA methylation, DNA methyltransferases (DNMTs), also directly repress transcription and associate with HDACs. Regulation of these above chromatin-DNA methylation interactions as a function of DNA replication timing is emerging as a key event in the inheritance of transcriptionally repressed domains of the genome. Importantly, synergy between HDAC activity and DNA methylation is operative for a key epigenetic abnormality in cancer cells, transcriptional silencing of tumor suppressor genes. This change has now been recognized for genes that are essential for normal regulation of virtually every major cell function including cell growth, differentiation, apoptosis, DNA repair, and cell-cell, cell-substratum interaction. Understanding the molecular determinants of both normal and abnormal patterns of chromatin formation and DNA methylation thus holds great promise for our understanding of cancer and for means to better diagnose, prevent, and treat this disease.


Subject(s)
Chromatin/genetics , DNA Methylation , Neoplasms/genetics , Animals , Chromatin/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/metabolism , Neoplasms/pathology , Transcription, Genetic
12.
J Biol Chem ; 276(34): 32282-7, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11427539

ABSTRACT

We demonstrate that the recently identified DNA methyltransferases, Dnmt3a and Dnmt3b, like DNMT1, repress transcription in a methylation-independent manner. Dnmt3a and Dnmt3b repress transcription primarily through a plant homeodomain-like motif that is shared with the ATRX protein but is not present in DNMT1. Unlike DNMT1, which localizes to replication foci during S-phase in murine embryonic fibroblasts, Dnmt3a co-localizes with heterochromatin protein 1 alpha (HP1 alpha) and methyl-CpG binding proteins throughout the cell cycle to late-replicating pericentromeric heterochromatin. In contrast to Dnmt3a, Dnmt3b remained diffuse in the nucleus of embryonic fibroblasts at all cell cycle stages. However, Dnmt3a and Dnmt3b co-localize to these pericentromeric heterochromatin regions in murine embryonic stem cells. This finding is important to the fact that mutations in DNMT3B are found in the developmental syndrome, ICF (immunodeficiency, centromeric heterochromatin instability, and facial anomalies), which involves extensive loss of methylation from pericentromeric regions. The localization of Dnmt3a and Dnmt3b was unaffected in Dnmt1 null embryonic stem cells, which lose the majority of methylation at pericentromeric major satellite repeats, suggesting that these enzymes are not dependent upon preexisting methylation for their targeting. DNMT1 is then positioned to reestablish transcriptionally repressive chromatin as cells replicate, while Dnmt3a and Dnmt3b may help to establish such chromatin in late S-phase and maintain this repressive heterochromatin throughout the cell cycle in a developmentally and/or cell type manner.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Helicases , Heterochromatin/metabolism , Nuclear Proteins , Repressor Proteins/metabolism , Transcription, Genetic , 3T3 Cells , Animals , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methyltransferase 3A , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Mice , Repressor Proteins/chemistry , Transcription Factors/metabolism , X-linked Nuclear Protein , DNA Methyltransferase 3B
13.
Cancer Res ; 61(12): 4689-92, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11406538

ABSTRACT

Defects in DNA repair may be responsible for the genesis of mutations in key genes in cancer cells. The tumor suppressor gene p53 is commonly mutated in human cancer by missense point mutations, most of them G:C to A:T transitions. A recognized cause for this type of change is spontaneous deamination of the methylcytosine. However, the persistence of a premutagenic O(6)-methylguanine can also be invoked. This last lesion is removed in the normal cell by the DNA repair enzyme O(6)-methylguanine-DNA methyltransferase (MGMT). In many tumor types, epigenetic silencing of MGMT by promoter hypermethylation has been demonstrated and linked to the appearance of G to A mutations in the K-ras oncogene in colorectal tumors. To study the relevance of defective MGMT function by aberrant methylation in relation to the presence of p53 mutations, we studied 314 colorectal tumors for MGMT promoter hypermethylation and p53 mutational spectrum. Inactivation of MGMT by aberrant methylation was associated with the appearance of G:C to A:T transition mutations at p53 (Fischer's exact test, two-tailed; P = 0.01). Overall, MGMT methylated tumors displayed p53 transition mutations in 43 of 126 (34%) cases, whereas MGMT unmethylated tumors only showed G:C to A:T changes in 37 of 188 (19%) tumors. A more striking association was found in G:C to A:T transitions in non-CpG dinucleotides; 71% (12 of 17) of the total non-CpG transition mutations in p53 were observed in MGMT aberrantly methylated tumors (Fischer's exact test, two-tailed; P = 0.008). Our data suggest that epigenetic silencing of MGMT by promoter hypermethylation may lead to G:C to A:T transition mutations in p53.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Genes, p53/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Point Mutation , Adenoma/genetics , Carcinoma/genetics , DNA Repair/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Promoter Regions, Genetic
15.
Cancer Res ; 61(7): 3200-5, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306509

ABSTRACT

Among the various forms of human lung cancer, small cell lung cancer (SCLC) exhibits a characteristic neuroendocrine (NE) phenotype. Neural and NE differentiation in SCLC depend, in part, on the action of the basic-helix-loop-helix (bHLH) transcription factor human achaete-scute homologue-1 (hASH1). In nervous system development, the Notch signaling pathway is a critical negative regulator of bHLH factors, including hASH1, controlling cell fate commitment and differentiation. To characterize Notch pathway function in SCLC, we explored the consequences of constitutively active Notch signaling in cultured SCLC cells. Recombinant adenoviruses were used to overexpress active forms of Notch1, Notch2, or the Notch effector protein human hairy enhancer of split-1 (HES1) in DMS53 and NCI-H209 SCLC cells. Notch proteins, but not HES1 or control adenoviruses, caused a profound growth arrest, associated with a G1 cell cycle block. We found up-regulation of p21(waf1/cip1) and p27kip1 in concert with the cell cycle changes. Active Notch proteins also led to dramatic reduction in hASH1 expression, as well as marked activation of phosphorylated extracellular signal-regulated kinase (ERK)1 and ERK2, findings that have been shown to be associated with cell cycle arrest in SCLC cells. These data suggest that the previously described function of Notch proteins as proto-oncogenes is highly context-dependent. Notch activation, in the setting of a highly proliferative hASH1-dependent NE neoplasm, can be associated with growth arrest and apparent reduction in neoplastic potential.


Subject(s)
Carcinoma, Small Cell/pathology , Cell Cycle Proteins , Homeodomain Proteins , Lung Neoplasms/pathology , Membrane Proteins/physiology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins , Basic Helix-Loop-Helix Transcription Factors , Cell Cycle/physiology , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/biosynthesis , Cyclins/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Enzyme Activation , G1 Phase/physiology , Helix-Loop-Helix Motifs , Humans , MAP Kinase Signaling System/physiology , Membrane Proteins/biosynthesis , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins/physiology , Receptor, Notch1 , Receptor, Notch2 , Receptors, Cell Surface/biosynthesis , Transcription Factor HES-1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation
16.
Cancer Res ; 61(8): 3225-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309270

ABSTRACT

We are in an era where the potential exists for deriving comprehensive profiles of DNA alterations characterizing each form of human cancer. Such profiles would provide invaluable insight into mechanisms underlying the evolution of each tumor type and will provide molecular markers, which could radically improve cancer detection. To date, no one type of DNA change has been defined which accomplishes this purpose. Herein, by using a candidate gene approach, we show that one category of DNA alteration, aberrant methylation of gene promoter regions, can enormously contribute to the above goals. We have now analyzed a series of promoter hypermethylation changes in 12 genes (p16(INK4a), p15(INK4b), p14(ARF), p73, APC,(5) BRCA1, hMLH1, GSTP1, MGMT, CDH1, TIMP3, and DAPK), each rigorously characterized for association with abnormal gene silencing in cancer, in DNA from over 600 primary tumor samples representing 15 major tumor types. The genes play known important roles in processes encompassing tumor suppression, cell cycle regulation, apoptosis, DNA repair, and metastastic potential. A unique profile of promoter hypermethylation exists for each human cancer in which some gene changes are shared and others are cancer-type specific. The hypermethylation of the genes occurs independently to the extent that a panel of three to four markers defines an abnormality in 70-90% of each cancer type. Our results provide an unusual view of the pervasiveness of DNA alterations, in this case an epigenetic change, in human cancer and a powerful set of markers to outline the disruption of critical pathways in tumorigenesis and for derivation of sensitive molecular detection strategies for virtually every human tumor type.


Subject(s)
DNA Methylation , Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Genetic Markers , Humans , Promoter Regions, Genetic
17.
Cancer Res ; 61(7): 2816-21, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306450

ABSTRACT

The INK4a/ARF locus encodes two distinct tumor suppressors, p16INK4a and p14ARF. Although the contribution of p16INK4a to human tumorigenesis through point mutation, deletion, and hypermethylation has been widely documented, little is known about specific p14ARF lesions and their consequences. Recent data indicate that p14ARF suffers inactivation by promoter hypermethylation in colorectal cancer cells. Because it is known that p14ARF prevents MDM2 nucleocytoplasmic shuttling and thus stabilizes p53 by attenuating MDM2-mediated degradation, we studied the relationship of p14ARF epigenetic silencing to the expression and localization of MDM2 and p53. Cancer cell lines with an unmethylated p14ARF promoter showed strong nuclear expression of MDM2, whereas in a colorectal cell line with p14ARF hypermethylation-associated inactivation, MDM2 protein was also seen in the cytosol. Treatment with the demethylating agent 5-aza-2'-deoxycytidine was able to reinternalize MDM2 to the nucleus, and p53 expression was restored. No apparent changes in retinoblastoma localization were observed. We also studied the profile of p14ARF promoter hypermethylation in an extensive collection of 559 human primary tumors of different cell types, observing that in colorectal, gastric, renal, esophageal, and endometrial neoplasms and gliomas, aberrant methylation of p14ARF was a relatively common epigenetic event. MDM2 expression patterns revealed that lack of p14ARF promoter hypermethylation was associated with tumors showing exclusive nuclear MDM2 staining, whereas MDM2 cytosolic staining was frequently observed in neoplasms with aberrant p14ARF methylation. Taken together, these data support that epigenetic silencing of p14ARF by promoter hypermethylation is a key mechanism in the disturbance of the MDM2 nuclear localization in human cancer.


Subject(s)
DNA Methylation , Gene Silencing , Nuclear Proteins , Promoter Regions, Genetic , Proteins/genetics , Proto-Oncogene Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Protein Biosynthesis , Proteins/physiology , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF
18.
Hum Mol Genet ; 10(7): 687-92, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11257100

ABSTRACT

Gene function in cancer can be disrupted either through genetic alterations, which directly mutate or delete genes, or epigenetic alterations, which alter the heritable state of gene expression. The latter events are mediated by formation of transcriptionally repressive chromatin states around gene transcription start sites and an associated gain of methylation in normally unmethylated CpG islands in these regions. The genes affected include over half of the tumor suppressor genes that cause familial cancers when mutated in the germline; the selective advantage for genetic and epigenetic dysfunction in these genes is very similar. The aberrant methylation can begin very early in tumor progression and mediate most of the important pathway abnormalities in cancer including loss of cell cycle control, altered function of transcription factors, altered receptor function, disruption of normal cell-cell and cell-substratum interaction, inactivation of signal transduction pathways, loss of apoptotic signals and genetic instability. The active role of the aberrant methylation in transcriptional silencing of genes is becoming increasingly understood and involves a synergy between the methylation and histone deacetylase (HDAC) activity. This synergy can be mediated directly by HDAC interaction with DNA methylating enzymes and by recruitment through complexes involving methyl-cytosine binding proteins. In the translational arena, the promoter hypermethylation changes hold great promise as DNA tumor markers and their potentially reversible state creates a target for cancer therapeutic strategies involving gene reactivation.


Subject(s)
Chromatin/physiology , DNA Methylation , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Animals , Chromatin/metabolism , CpG Islands , Disease Progression , Gene Silencing , Humans , Models, Biological , Mutation , Promoter Regions, Genetic , Transcription, Genetic
19.
J Biol Chem ; 276(2): 1634-42, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11038348

ABSTRACT

p16(INK4a) is frequently altered in human cancer, often through epigenetically mediated transcriptional silencing. However, little is known about the transcriptional regulation of this gene. To learn more about such control, we initiated studies of proteins that bind to the promoter in cancer cells that do, and do not, express the gene. We identify RNA helicase A (RHA) as a protein that binds much better to the p16(INK4a) promoter in the expressing cells. RHA has not previously been characterized to manifest sequence-specific DNA interaction but does so to the sequence 5' CGG ACC GCG TGC GC 3' in the p16(INK4a) promoter. The Drosophila homologue to RHA, maleless (Mle), functions in the fly for 2-fold activation of male X-chromosome genes. In our experimental setting, RHA induces a similar modest up-regulation of the p16(INK4a) promoter that is dependent upon its sequence-specific interaction. Mle colocalizes with hyperacetylated H4Ac16 on the X-chromosome and some autosomal loci. The decreased binding of RHA to p16(INK4a) in our cells, where the gene is transcriptionally inactive, is associated with decreased amounts of RHA that immunoprecipitate with acetylated lysine antibodies. Finally, we show RHA to be a cellular substrate for caspase-3, which decreases its sequence-specific binding to p16(INK4a) by cleavage of the N terminus. Thus, we have identified a new protein interaction with the p16(INK4a) promoter that involves an important protein for transcriptional modulation. This interaction is decreased in cancer cells, where this gene is aberrantly transcriptionally silent.


Subject(s)
Autoantigens/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genes, Tumor Suppressor , Promoter Regions, Genetic , RNA Helicases/metabolism , Animals , Autoantigens/genetics , Base Sequence , Binding Sites , Carcinoma, Small Cell , Carcinoma, Squamous Cell , Cell Nucleus/metabolism , DEAD-box RNA Helicases , DNA-Binding Proteins/metabolism , Drosophila/genetics , Gene Expression Regulation , Humans , Lung Neoplasms , Male , Neoplasm Proteins , Protein Biosynthesis , RNA Helicases/genetics , Rabbits , Recombinant Proteins/metabolism , Reticulocytes/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription, Genetic , Transfection , Tumor Cells, Cultured , X Chromosome
20.
Curr Protoc Hum Genet ; Chapter 10: Unit 10.6, 2001 May.
Article in English | MEDLINE | ID: mdl-18428243

ABSTRACT

Methylation-specific PCR is a rapid method used to determine the methylation status of DNA. Not only does methylation affect the expression of genes in normal cells, but it is now known that disease processes such as cancer can result in abnormal DNA methylation patterns. Methylation-specific PCR can be used to investigate imprinted genes, to assess human tumors for clonality by studying genes inactivated on the X chromosome, and to examine abnormally methylated CpG islands in neoplasia.


Subject(s)
DNA Methylation , Polymerase Chain Reaction/methods , CpG Islands , Female , Genetics, Medical , Genomic Imprinting , Humans , Neoplasms/genetics , X Chromosome Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL
...