Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
1.
BMC Cancer ; 23(1): 356, 2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37072777

ABSTRACT

Colorectal cancer (CRC) is the third most prevalent cancer in the world, yet the sensitivity and specificity of biomarkers for CRC diagnosis are insufficient. In the present study, we performed a protein microarray screening method to identify antibody markers for CRC. Inhibitor of growth family 1 (ING1) was identified as a candidate tumor antigen for CRC using protein microarrays (ProtoArray). Subsequent amplified luminescence proximity homogeneous assay-linked immunosorbent assay using recombinant ING1 protein showed that the serum levels of anti-ING1 antibodies were increased not only in patients with CRC but also in those with esophageal cancer (EC), gastric cancer (GC), breast cancer (BrC), and pancreatic cancer (PC) compared with those of healthy donors (HDs). Antibodies against the ING1 amino acids between 239 and 253 were present at significantly higher levels in patients with CRC than in those with EC, GC, BrC, or PC. Anti-ING1 antibody levels were significantly higher in the patients with CRC at any stages than in the HDs. Immunohistochemical staining revealed higher expression of ING1 protein in CRC cells than in the adjacent normal tissues. In luciferase reporter assays using a CRC cell line, ING1 augmented p53-mediated NOXA promoter activity but attenuated p53-stimulated Bax, p21, and PUMA promoter activities. Consequently, serum anti-ING1 antibodies can be used for sensitive and specific diagnoses of CRC.


Subject(s)
Colorectal Neoplasms , Tumor Suppressor Proteins , Humans , Inhibitor of Growth Protein 1/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Nuclear Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Autoantibodies , Colorectal Neoplasms/diagnosis
2.
Genome Med ; 13(1): 158, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34635154

ABSTRACT

BACKGROUND: Enhancers are distal cis-regulatory elements required for cell-specific gene expression and cell fate determination. In cancer, enhancer variation has been proposed as a major cause of inter-patient heterogeneity-however, most predicted enhancer regions remain to be functionally tested. METHODS: We analyzed 132 epigenomic histone modification profiles of 18 primary gastric cancer (GC) samples, 18 normal gastric tissues, and 28 GC cell lines using Nano-ChIP-seq technology. We applied Capture-based Self-Transcribing Active Regulatory Region sequencing (CapSTARR-seq) to assess functional enhancer activity. An Activity-by-contact (ABC) model was employed to explore the effects of histone acetylation and CapSTARR-seq levels on enhancer-promoter interactions. RESULTS: We report a comprehensive catalog of 75,730 recurrent predicted enhancers, the majority of which are GC-associated in vivo (> 50,000) and associated with lower somatic mutation rates inferred by whole-genome sequencing. Applying CapSTARR-seq to the enhancer catalog, we observed significant correlations between CapSTARR-seq functional activity and H3K27ac/H3K4me1 levels. Super-enhancer regions exhibited increased CapSTARR-seq signals compared to regular enhancers, even when decoupled from native chromatin contexture. We show that combining histone modification and CapSTARR-seq functional enhancer data improves the prediction of enhancer-promoter interactions and pinpointing of germline single nucleotide polymorphisms (SNPs), somatic copy number alterations (SCNAs), and trans-acting TFs involved in GC expression. We identified cancer-relevant genes (ING1, ARL4C) whose expression between patients is influenced by enhancer differences in genomic copy number and germline SNPs, and HNF4α as a master trans-acting factor associated with GC enhancer heterogeneity. CONCLUSIONS: Our results indicate that combining histone modification and functional assay data may provide a more accurate metric to assess enhancer activity than either platform individually, providing insights into the relative contribution of genetic (cis) and regulatory (trans) mechanisms to GC enhancer functional heterogeneity.


Subject(s)
Enhancer Elements, Genetic , Epigenomics , Stomach Neoplasms/genetics , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Acetylation , Cell Line, Tumor , Cell Proliferation , Chromatin , Gene Expression Regulation, Neoplastic , Genomics , Histones/metabolism , Humans , Inhibitor of Growth Protein 1/genetics , Inhibitor of Growth Protein 1/metabolism , Oncogenes , Promoter Regions, Genetic , RNA-Seq , Transcriptome , Whole Genome Sequencing
3.
Open Biol ; 11(9): 210077, 2021 09.
Article in English | MEDLINE | ID: mdl-34493070

ABSTRACT

Approximately 90% of cancer-related deaths can be attributed to a tumour's ability to spread. We have identified CG7379, the fly orthologue of human ING1, as a potent invasion suppressor. ING1 is a type II tumour suppressor with well-established roles in the transcriptional regulation of genes that control cell proliferation, response to DNA damage, oncogene-induced senescence and apoptosis. Recent work suggests a possible role for ING1 in cancer cell invasion and metastasis, but the molecular mechanism underlying this observation is lacking. Our results show that reduced expression of CG7379 promotes invasion in vivo in Drosophila, reduces the junctional localization of several adherens and septate junction components, and severely disrupts cell-cell junction architecture. Similarly, ING1 knockdown significantly enhances invasion in vitro and disrupts E-cadherin distribution at cell-cell junctions. A transcriptome analysis reveals that loss of ING1 affects the expression of several junctional and cytoskeletal modulators, confirming ING1 as an invasion suppressor and a key regulator of cell-cell junction integrity.


Subject(s)
Breast Neoplasms/prevention & control , Cell Communication , Drosophila Proteins/metabolism , Gene Expression Regulation, Neoplastic , Inhibitor of Growth Protein 1/metabolism , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Humans , Inhibitor of Growth Protein 1/genetics , MCF-7 Cells , Neoplasm Invasiveness , Transcriptome
4.
Curr Genet ; 67(4): 535-538, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33641041

ABSTRACT

The histone modifications usually form complicated networks to regulate accessibility of DNA and transcription. Identification of proteins that are involved in the crosstalk among different histone modifications will help to better understand the epigenetic regulatory network in eukaryotes. The Inhibitor of Growth (ING) proteins represent a tumor suppressor family were first linked to histone modification in yeast and their functions in epigenetic regulation were further characterized. This review summarizes the crosstalk of histone modification in fungi and describes recently achieved mechanistic insights into the role of Fng1 (an ING protein in filamentous ascomycetes) in this process. We conclude that Fng1 is involved in crosstalk among histone acetylation, deacetylation and methylation.


Subject(s)
Epigenesis, Genetic/genetics , Histone Code/genetics , Inhibitor of Growth Protein 1/genetics , Protein Processing, Post-Translational/genetics , Acetylation , Fusarium/genetics , Gene Expression Regulation, Fungal/genetics , Histone Acetyltransferases/genetics , Methylation , Schizosaccharomyces/genetics
6.
Trends Genet ; 36(2): 71-80, 2020 02.
Article in English | MEDLINE | ID: mdl-31822355

ABSTRACT

Changes in epigenetic DNA methylation are the most promising predictor of biological age and lifespan in humans, but whether methylation changes affect ageing is unresolved. Here, we discuss converging data, which indicate that one mode by which aberrant DNA methylation can affect ageing is via CCAAT/enhancer binding protein beta (C/EBPß). This basic leucine-zipper (bZIP) transcription factor is controlled by the lifespan regulator mechanistic/mammalian target of rapamycin complex 1 (mTORC1) and plays an important role in energy homeostasis and adipose tissue differentiation. Emerging evidence indicates that access of C/EBPß proteins to cognate binding sites is regulated by DNA demethylation via ten-eleven translocation (TET) methylcytosine dioxygenases and their adaptor proteins growth arrest and DNA damage-inducible protein 45 alpha (GADD45α) and inhibitor of growth 1 (ING1). We discuss the emerging causal nexus between C/EBPß, energy metabolism, and DNA demethylation in organismal ageing.


Subject(s)
Aging/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Cycle Proteins/genetics , DNA Methylation/genetics , Inhibitor of Growth Protein 1/genetics , Aging/pathology , Cell Differentiation/genetics , Energy Metabolism/genetics , Epigenesis, Genetic/genetics , Humans
7.
Cancer Lett ; 447: 66-74, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30673590

ABSTRACT

ING family genes (Inhibitor of Growth) are tumor suppressor genes that play a vital role in cell homeostasis. It has been shown that their expression is lost or diminished in many cancers and other diseases. The main mechanisms by which they are regulated in oncogenesis have not yet been fully elucidated. The involvement of non-coding RNAs (ncRNAs) and in particular microRNAs (miRNAs) in post-transcriptional gene regulation is well established. miRNAs are short sequences (18-25 nucleotides) that can bind to the 3 'UTR sequence of the targeted messenger RNA (mRNA), leading to its degradation or translational repression. Interactions between the ING family and miRNAs have been described in some cancers but also in other diseases. The involvement of miRNAs in ING family regulation opens up new fields of investigation, particularly for targeted therapies. In this review, we will summarize the regulatory mechanisms at the RNA and protein level of the ING family and focus on the interactions with ncRNAs.


Subject(s)
Disease/genetics , Inhibitor of Growth Protein 1/genetics , Neoplasms/genetics , RNA, Untranslated/genetics , Animals , Gene Expression Regulation/genetics , Humans , MicroRNAs/genetics , RNA, Messenger/genetics
8.
Mech Ageing Dev ; 177: 109-117, 2019 01.
Article in English | MEDLINE | ID: mdl-29928906

ABSTRACT

Replicative capacity of normal human cells decreases as telomeric sequence is lost at each division. It is believed that when a subset of chromosomes reach a critically short length, an ATM-initiated and p53-mediated transcriptional response inhibits cell growth, promoting cell senescence. In addition to loss of telomeric sequence, senescence can be induced by other stresses including ionizing radiation, oxidative damage, chemical crosslinkers like the chemotherapeutic agent cisplatin, as well as overactivation of oncogenes and tumor suppressors. Our group found that the expression of an isoform of the INhibitor of Growth 1 gene called ING1a increases approximately 10-fold as fibroblasts approach senescence and that forced expression rapidly induces a senescent phenotype in primary diploid fibroblasts, epithelial and endothelial cells that resembles replicative senescence by most physical and biochemical measures. ING1a induces these changes through strongly inhibiting endocytosis to block mitogen signaling by inducing the expression of intersectin 2, a key scaffolding protein of the endosomal pathway. This, in turn increases the expression of Rb and of p57Kip2 and p16INK4a that serve to maintain Rb is an active, growth inhibitory state. The ING1a model is currently being used to better understand the mechanism(s) responsible for activating Rb to enforce the senescent state.


Subject(s)
Cellular Senescence/physiology , Gene Expression Regulation/physiology , Inhibitor of Growth Protein 1/biosynthesis , Models, Biological , Adaptor Proteins, Vesicular Transport/biosynthesis , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/genetics , Endocytosis/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/pathology , Humans , Inhibitor of Growth Protein 1/genetics , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
9.
Cell Death Dis ; 9(10): 1032, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30302016

ABSTRACT

Glioblastoma is the most common and aggressive primary brain tumor in adults. New drug design and development is still a major challenge for glioma treatment. Increasing evidence has shown that nitazoxanide, an antiprotozoal drug, has a novel antitumor role in various tumors and exhibits multiple molecular functions, especially autophagic regulation. However, whether nitazoxanide-associated autophagy has an antineoplastic effect in glioma remains unclear. Here, we aimed to explore the underlying molecular mechanism of nitazoxanide in glioblastoma. Our results showed that nitazoxanide suppressed cell growth and induced cell cycle arrest in glioblastoma by upregulating ING1 expression with a favorable toxicity profile. Nitazoxanide inhibited autophagy through blockage of late-stage lysosome acidification, resulting in decreased cleavage of ING1. A combination with chloroquine or Torin1 enhanced or impaired the chemotherapeutic effect of nitazoxanide in glioblastoma cells. Taken together, these findings indicate that nitazoxanide as an autophagy inhibitor induces cell cycle arrest in glioblastoma via upregulated ING1 due to increased transcription and decreased post-translational degradation by late-stage autophagic inhibition.


Subject(s)
Antiprotozoal Agents/pharmacology , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Glioblastoma/drug therapy , Glioblastoma/metabolism , Inhibitor of Growth Protein 1/metabolism , Thiazoles/pharmacology , Animals , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/pharmacology , Glioma/drug therapy , Glioma/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Naphthyridines/pharmacology , Nitro Compounds , Protein Processing, Post-Translational/drug effects , Transcription, Genetic/drug effects , Up-Regulation/drug effects
10.
Biomed Pharmacother ; 108: 1628-1639, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30372865

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) have been consistently demonstrated to be involved in non-small cell lung cancer (NSCLC) as either tumor oncogenes or tumor suppressors. However, the detailed role of miR-500 and miR-628 in NSCLC remain poorly understood. METHODS: The expressions of miR-500 and miR-628 in NSCLC tissues and cell lines were measured by quantitative real-time PCR (qRT-PCR). Cells migration, invasion, proliferation, adhesion and apoptosis abilities were test to analyze the biological functions of miR-500 and miR-628 in NSCLC. A bioinformatic analysis was conducted to predict the target genes regulated by miR-500 and miR-628 using TargetScan (http://www.targetscan.org/mamm/). Luciferase reporter assay was employed to validate the direct targeting of ING1 by miR-500 and miR-628. RESULTS: In this study, miR-500 and miR-628 were up-regulated with NSCLC tissues. Furthermore, inhibition of miR-500 and miR-628 significantly suppressed NSCLC cells proliferation, migration, invasion and adhesion, and induced NSCLC cells apoptosis. Additionally, the result showed that ING1 functioned as the direct target for miR-500 and miR-628, which was a core tumor suppressor in regulating NSCLC progression. Over-expression of ING1 could dramatically inhibit NSCLC cells proliferation, migration and invasion, and promote cells apoptosis. CONCLUSION: These results brought new insights into the oncogenic role of miR-500 and miR-628 in NSCLC, indicating that miR-500 and miR-628 might be the novel biomarkers for the diagnosis and prognosis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement/physiology , Inhibitor of Growth Protein 1/biosynthesis , Lung Neoplasms/metabolism , MicroRNAs/metabolism , A549 Cells , Carcinoma, Non-Small-Cell Lung/prevention & control , Cell Proliferation/physiology , Down-Regulation/physiology , Gene Targeting/methods , Humans , Inhibitor of Growth Protein 1/antagonists & inhibitors , Lung Neoplasms/prevention & control , MicroRNAs/antagonists & inhibitors
11.
Genes Dev ; 32(11-12): 742-762, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29884649

ABSTRACT

Changes in DNA methylation are among the best-documented epigenetic alterations accompanying organismal aging. However, whether and how altered DNA methylation is causally involved in aging have remained elusive. GADD45α (growth arrest and DNA damage protein 45A) and ING1 (inhibitor of growth family member 1) are adapter proteins for site-specific demethylation by TET (ten-eleven translocation) methylcytosine dioxygenases. Here we show that Gadd45a/Ing1 double-knockout mice display segmental progeria and phenocopy impaired energy homeostasis and lipodystrophy characteristic of Cebp (CCAAT/enhancer-binding protein) mutants. Correspondingly, GADD45α occupies C/EBPß/δ-dependent superenhancers and, cooperatively with ING1, promotes local DNA demethylation via long-range chromatin loops to permit C/EBPß recruitment. The results indicate that enhancer methylation can affect aging and imply that C/EBP proteins play an unexpected role in this process. Our study suggests a causal nexus between DNA demethylation, metabolism, and organismal aging.


Subject(s)
Aging, Premature/genetics , Aging/genetics , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Demethylation , Inhibitor of Growth Protein 1/metabolism , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Homeostasis/genetics , Inhibitor of Growth Protein 1/genetics , Lipodystrophy/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics
12.
Neuroscience ; 369: 248-260, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29158107

ABSTRACT

Epigenetic regulation of activity-induced gene expression involves multiple levels of molecular interaction, including histone and DNA modifications, as well as mechanisms of DNA repair. Here we demonstrate that the genome-wide deposition of inhibitor of growth family member 1 (ING1), which is a central epigenetic regulatory protein, is dynamically regulated in response to activity in primary cortical neurons. ING1 knockdown leads to decreased expression of genes related to synaptic plasticity, including the regulatory subunit of calcineurin, Ppp3r1. In addition, ING1 binding at a site upstream of the transcription start site (TSS) of Ppp3r1 depends on yet another group of neuroepigenetic regulatory proteins, the Piwi-like family, which are also involved in DNA repair. These findings provide new insight into a novel mode of activity-induced gene expression, which involves the interaction between different epigenetic regulatory mechanisms traditionally associated with gene repression and DNA repair.


Subject(s)
Cerebral Cortex/metabolism , Epigenesis, Genetic , Inhibitor of Growth Protein 1/metabolism , Neurons/metabolism , Animals , Argonaute Proteins/metabolism , Calcium-Binding Proteins , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Inhibitor of Growth Protein 1/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Muscle Proteins/metabolism , RNA, Messenger/metabolism
13.
Mol Med Rep ; 16(5): 7367-7374, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28944862

ABSTRACT

Inhibitor of growth 2 (ING2) is involved in chromatin remodeling and it has previously been suggested that ING2 may regulate gene expression. The authors previously identified matrix metalloproteinase 13 (MMP13) as a target gene of ING2 in colorectal cancer. The aim of the present study was to identify novel genes regulated by ING2 and histone deacetylase 1 (HDAC1) and to clarify the biological significance of the ING2 structure. The present study generated the point mutant constructs of ING2 and deletion constructs consisting of partial ING2 to investigate the effect on gene expression and verify the interaction with HDAC1, mSin3A and sap30. A microarray was performed to find novel ING2/HDAC1 target genes using cell co­overexpression of ING2 and HDAC1. Plasminogen activator inhibitor­1 (PAI­1) was upregulated with overexpression of ING1b and ING2. The mutation of the PHD domain at 218 significantly attenuated the MMP13 and PAI­1 expression, whereas the mutation at 224 resulted in increased expression. Furthermore, the expression levels were slightly reduced by the mutation of the C­terminal. The lack of the PHD domain and the C­terminal in ING2 resulted in a decreased ability to induce gene expression. The C­terminal with PHD domain, which lacked the N­terminal, maintained the transactive function for regulating the target genes. In addition to MMP13 and PAI­1, eight genes [heat shock protein family A member 1A (HSPA1A), MIR7­3 host gene, chorionic somatomammotropin hormone 1, growth arrest and DNA damage inducible b, dehydrogenase/reductase 2, galectin 1, myosin light chain 1, and VGF nerve growth factor inducible] were demonstrated to be associated with ING2/HDAC1. The present study demonstrated that ING2/HDAC1 regulated PAI­1 and HSPA1A expression and the PHD domain and the C­terminal of ING2, which are binding sites of HDAC1 and mSin3A, are essential regions for the regulation of gene expression.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Histone Deacetylase 1/metabolism , Homeodomain Proteins/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , Histone Deacetylase 1/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Inhibitor of Growth Protein 1/chemistry , Inhibitor of Growth Protein 1/genetics , Inhibitor of Growth Protein 1/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 2/metabolism , PHD Zinc Fingers , Plasmids/genetics , Plasmids/metabolism , Plasminogen Activator Inhibitor 1/genetics , Point Mutation , Protein Array Analysis , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Sin3 Histone Deacetylase and Corepressor Complex , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Up-Regulation
14.
Aging Cell ; 16(5): 918-933, 2017 10.
Article in English | MEDLINE | ID: mdl-28703423

ABSTRACT

Deregulation of precursor mRNA splicing is associated with many illnesses and has been linked to age-related chronic diseases. Here we review recent progress documenting how defects in the machinery that performs intron removal and controls splice site selection contribute to cellular senescence and organismal aging. We discuss the functional association linking p53, IGF-1, SIRT1, and ING-1 splice variants with senescence and aging, and review a selection of splicing defects occurring in accelerated aging (progeria), vascular aging, and Alzheimer's disease. Overall, it is becoming increasingly clear that changes in the activity of splicing factors and in the production of key splice variants can impact cellular senescence and the aging phenotype.


Subject(s)
Aging/genetics , Alternative Splicing , Alzheimer Disease/genetics , Progeria/genetics , RNA Precursors/genetics , RNA, Messenger/genetics , Aging/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Cellular Senescence/genetics , Exons , Gene Expression Regulation, Developmental , Humans , Inhibitor of Growth Protein 1/genetics , Inhibitor of Growth Protein 1/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Introns , Progeria/metabolism , Progeria/physiopathology , RNA Precursors/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA, Messenger/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Nucleic Acids Res ; 45(4): 1776-1792, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27903908

ABSTRACT

Epigenetic, transcriptional and signaling processes in the nucleolus regulate rRNA transcription and cell growth. We report here that the tumor suppressor ING1b binds rDNA, regulates rDNA chromatin modifications and affects nucleolar localization of mTOR to modulate rRNA levels. ING1 represses rDNA transcription by recruiting HDAC1 to rDNA loci, increasing its association with the NoRC complex and deacetylating the histone H3K9 and H3K27 marks of active transcription. Loss of ING1 enhances nucleolar localization of phospho-mTOR and its association with Raptor and GßL, even during rapamycin treatment. ING1 inhibits rDNA transcription by inhibiting UBF activity and its interaction with mTOR. Regulation of rDNA heterochromatin and rRNA synthesis by ING1 is also apparent during normal cell growth and during cell stress. Moreover, this function was also important during PMA induced differentiation of THP1 cells, since knocking down ING1 affected the process by inhibiting rRNA transcriptional repression. These observations show that ING1 regulates the nucleolar epigenome and rDNA transcription suggesting that regulation of protein synthesis might serve as the basis for ING1 function as a type II tumor suppressor.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Ribosomal/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Cell Differentiation/genetics , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Silencing , Genes, Tumor Suppressor , Glucose/metabolism , Histone Deacetylase 1/metabolism , Humans , Inhibitor of Growth Protein 1 , Monocytes/cytology , Monocytes/metabolism , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , RNA Precursors/genetics , RNA Precursors/metabolism
16.
J Mol Med (Berl) ; 94(10): 1167-1179, 2016 10.
Article in English | MEDLINE | ID: mdl-27305909

ABSTRACT

The androgen receptor (AR) is a transcriptional factor that has a pivotal role in the development of normal and also cancerous prostate. Therefore, analyzing AR signaling is essential to understand cancerogensis and proliferation of prostate cancer (PCa). Inhibitor of growth 1 (ING1) and ING2 are tumor suppressors with reduced expression in many cancer types. There are also indications of misregulation of ING1 and ING2 in PCa. However, the roles of ING1 and ING2 in PCa and AR signaling are poorly understood. Here, we show that surprisingly the ING1b knockdown (KD) represses AR-mediated transactivation on AR key target genes in the human LNCaP PCa cells. This is associated with growth reduction of LNCaP cells by ING1 KD. In line with this, using Ing1 knockout (KO) mice, we provide further evidence that ING1 deficiency downregulates prostate-specific AR target genes in vivo. Further analyses suggest that KD of ING1b results in induction of both cellular senescence and the cell cycle inhibitor p16 INK4a . The unexpected finding that the ING1 KD results in growth inhibition was further analyzed and can be explained by a compensatory mechanism through enhanced levels of ING2 protein in ING1-deficient condition. Accordingly, the data suggest that ING2 interacts with AR and hampers the AR transcriptional activation, causes growth arrest, and induces cellular senescence. The data further suggest that ING2 upregulates p16 INK4a , which is a novel target for ING2. Taken together, our data suggest that ING2 is a novel corepressor for AR. ING2 levels are increased upon downregulation of ING1 expression indicating a compensatory mechanism and suggests a novel crosstalk between ING1 and ING2 tumor suppressors to inhibit AR signaling and induce cellular senescence in PCa cells. KEY MESSAGE: • The tumor suppressors ING1 and 2 are dysregulated in human prostate cancer. • ING1 deficiency reduces AR-mediated gene expression in vitro and in vivo. • ING2, like ING1, inhibits AR-mediated transactivation and prostate cancer cell growth. • ING1 regulates ING2. • ING1 and ING2 crosstalk with each other to inhibit AR signaling in prostate cancer.


Subject(s)
Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Androgen/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cellular Senescence , Humans , Inhibitor of Growth Protein 1 , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice, Knockout , Nuclear Proteins/genetics , Prostate/metabolism , Prostatic Neoplasms/metabolism , Seminal Vesicles/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics
17.
J Mol Cell Biol ; 8(3): 207-20, 2016 06.
Article in English | MEDLINE | ID: mdl-26993046

ABSTRACT

The androgen receptor (AR) signaling is critical for prostate cancer (PCa) progression to the castration-resistant stage with poor clinical outcome. Altered function of AR-interacting factors may contribute to castration-resistant PCa (CRPCa). Inhibitor of growth 1 (ING1) is a tumor suppressor that regulates various cellular processes including cell proliferation. Interestingly, ING1 expression is upregulated in senescent primary human prostate cells; however, its role in AR signaling in PCa was unknown. Using a proteomic approach by surface-enhanced laser desorption ionization-mass spectrometry (SELDI-MS) combined with immunological techniques, we provide here evidence that ING1b interacts in vivo with the AR. The interaction was confirmed by co-immunoprecipitation, in vitro GST-pull-down, and quantitative intracellular colocalization analyses. Functionally, ING1b inhibits AR-responsive promoters and endogenous key AR target genes in the human PCa LNCaP cells. Conversely, ING1b knockout (KO) mouse embryonic fibroblasts (MEFs) exhibit enhanced AR activity, suggesting that the interaction with ING1b represses the AR-mediated transcription. Also, data suggest that ING1b expression is downregulated in CRPCa cells compared with androgen-dependent LNCaP cells. Interestingly, its ectopic expression induces cellular senescence and reduces cell migration in both androgen-dependent and CRPCa cells. Intriguingly, ING1b can also inhibit androgen-induced growth in LNCaP cells in a similar manner as AR antagonists. Moreover, ING1b upregulates different cell cycle inhibitors including p27(KIP1), which is a novel target for ING1b. Taken together, our findings reveal a novel corepressor function of ING1b on various AR functions, thereby inhibiting PCa cell growth.


Subject(s)
Cellular Senescence , Co-Repressor Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Inhibitor of Growth Protein 1 , Male , Mice , NIH 3T3 Cells , Prostatic Neoplasms/genetics , Protein Binding , Transcription, Genetic , Transcriptional Activation/genetics
18.
Ukr Biochem J ; 88(1): 11-21, 2016.
Article in English | MEDLINE | ID: mdl-29227074

ABSTRACT

We have studied the effect of inhibition of IRE1 (inositol requiring enzyme 1), which is a central mediator of endoplasmic reticulum stress and a controller of cell proliferation and tumor growth, on hypoxic regulation of the expression of different proliferation related genes in U87 glioma cells. It was shown that hypoxia leads to up-regulation of the expression of IL13RA2, CD24, ING1, ING2, ENDOG, and POLG genes and to down-regulation ­ of KRT18, TRAPPC3, TSFM, and MTIF2 genes at the mRNA level in control glioma cells. Changes for ING1 and CD24 genes were more significant. At the same time, inhibition of IRE1 modifies the effect of hypoxia on the expression of all studied genes. In particular, it increases sensitivity to hypoxia of the expression of IL13RA2, TRAPPC3, ENDOG, and PLOG genes and suppresses the effect of hypoxia on the expression of ING1 gene. Additionally, it eliminates hypoxic regulation of KRT18, CD24, ING2, TSFM, and MTIF2 genes expressions and introduces sensitivity to hypoxia of the expression of BET1 gene in glioma cells. The present study demonstrates that hypoxia, which often contributes to tumor growth, affects the expression of almost all studied genes. Additionally, inhibition of IRE1 can both enhance and suppress the hypoxic regulation of these gene expressions in a gene specific manner and thus possibly contributes to slower glioma growth, but several aspects of this regulation must be further clarified.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Neuroglia/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Signal Transduction/genetics , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , DNA Polymerase gamma/genetics , DNA Polymerase gamma/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Inhibitor of Growth Protein 1/genetics , Inhibitor of Growth Protein 1/metabolism , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-13 Receptor alpha2 Subunit/metabolism , Keratin-18/genetics , Keratin-18/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neuroglia/pathology , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
19.
Oncotarget ; 6(33): 34118-27, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26439691

ABSTRACT

Cell senescence contributes to organismal aging and is induced by telomere erosion and an ensuing DNA damage signal as cells reach the end of their replicative lifespan in vitro or in vivo. Stresses induced by oncogene or tumor suppressor hyperactivation, oxidative stress, ionizing radiation and other DNA damaging agents result in forms of stress induced premature senescence (SIPS) that show similarities to replicative senescence. Since replicative senescence and SIPS occur over many days and many population doublings of the mass cultures of primary cells used to study senescence, the sequence of events that occur downstream of senescence signaling can be challenging to define. Here we compare a new model of ING1a-induced senescence with several other forms of senescence. The ING1a epigenetic regulator synchronously induces senescence in mass cultures several-fold faster than all other agents, taking 24 and 36 hours to activate the Rb/ p16INK4a, but not the p53 tumor suppressor axis to efficiently induce senescence. ING1a induces expression of intersectin 2, a scaffold protein necessary for endocytosis, altering the stoichiometry of endocytosis proteins, subsequently blocking growth factor uptake leading to activation of Rb signaling to block cell growth. ING1a acts as a novel link in the activation of the Rb pathway that can impose senescence in the absence of activating p53-mediated DNA damage signaling, and should prove useful in defining the molecular events contributing to Rb-induced senescence.


Subject(s)
Aging, Premature/genetics , Cellular Senescence/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Retinoblastoma Protein/metabolism , Stress, Physiological/genetics , Tumor Suppressor Proteins/genetics , Actin Cytoskeleton/metabolism , Adaptor Proteins, Vesicular Transport/biosynthesis , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/genetics , Endocytosis/physiology , Endothelial Cells/metabolism , Humans , Inhibitor of Growth Protein 1 , Keratinocytes/metabolism , Telomere Homeostasis/genetics , Tumor Suppressor Protein p53/metabolism
20.
Mol Cancer ; 14: 164, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26306560

ABSTRACT

BACKGROUND: Previous studies have established that levels of the Inhibitor of Growth 1(ING1) tumor suppressor are reduced in a significant proportion of different cancer types. Here we analyzed levels of ING1 in breast cancer patients to determine its prognostic significance as a biomarker for breast cancer prognosis. METHODS: We used automated quantitative analysis (AQUA) to determine the levels of ING1 in the tumor associated stromal cells of 462 breast cancer samples. To better understand how high ING1 levels affect nearby epithelium, we measured the levels of cytokines and secreted matrix metalloproteases (MMPs), using an ELISA based assay in mammary fibroblasts overexpressing ING1. These cells were also used in a 3-dimensional co-culture with MCF7 cells to determine the effect of released MMPs and other cytokines on growing colonies. RESULTS: We find that high levels of ING1 in stroma are associated with tumor grade (p = 0.001) and size (p = 0.02), and inversely associated with patient survival (p = 0.0001) in luminal, but not in non-luminal cancers, suggesting that high stromal ING1 promotes cancer development. In this group of patients ING1 could also predict patient survival and act as a biomarker (HR = 2.125). While ING1 increased or decreased the expression of different cytokines, ING1 also increased the levels of MMP1, MMP3 and MMP10 by 5-8 fold, and concomitantly decreased levels of the tissue inhibitors of metalloproteases TIMP2, TIMP3 and TIMP4 by 1.5-3.3 fold, resulting in significant increases in MMP activity as determined by zymography. Co-culturing of MCF7 cells with stromal cells expressing ING1 in 3-dimensional organoid cultures suggested that MCF7 colonies were less well defined, suggesting that secreted MMPs might promote migration. CONCLUSION: These data indicate that stromal ING1 expression can predict the survival of patients with luminal breast cancer. High levels of ING1 in stromal cells can promote the development of breast cancer through increased expression and release of MMPs and down regulation of TIMPs, which may be an underlying mechanism of reduced patient survival.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/genetics , Intracellular Signaling Peptides and Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Prognosis , Tumor Suppressor Proteins/biosynthesis , Adult , Aged , Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Disease-Free Survival , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Growth Protein 1 , Intracellular Signaling Peptides and Proteins/genetics , MCF-7 Cells , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Middle Aged , Nuclear Proteins/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...