Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Cell Biochem ; 125(6): e30570, 2024 06.
Article in English | MEDLINE | ID: mdl-38616697

ABSTRACT

Runt-related transcription factor 1 (RUNX1) plays an important role in normal haematopoietic cell development and function, and its function is frequently disrupted in leukaemia. RUNX1 is widely recognised as a sequence-specific DNA binding factor that recognises the motif 5'-TG(T/C)GGT-3' in promoter and enhancer regions of its target genes. Moreover, RUNX1 fusion proteins, such as RUNX1-ETO formed by the t(8;21) translocation, retain the ability to recognise and bind to this sequence to elicit atypical gene regulatory effects on bona fide RUNX1 targets. However, our analysis of publicly available RUNX1 chromatin immunoprecipitation sequencing (ChIP-Seq) data has provided evidence challenging this dogma, revealing that this motif-specific model of RUNX1 recruitment and function is incomplete. Our analyses revealed that the majority of RUNX1 genomic localisation occurs outside of promoters, that 20% of RUNX1 binding sites lack consensus RUNX motifs, and that binding in the absence of a cognate binding site is more common in promoter regions compared to distal sites. Reporter assays demonstrate that RUNX1 can drive promoter activity in the absence of a recognised DNA binding motif, in contrast to RUNX1-ETO. RUNX1-ETO supresses activity when it is recruited to promoters containing a sequence specific motif, while interestingly, it binds but does not repress promoters devoid of a RUNX1 recognition site. These data suggest that RUNX1 regulation of target genes occurs through multiple mechanisms depending on genomic location, the type of regulatory element and mode of recruitment.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Promoter Regions, Genetic , Humans , Binding Sites , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , DNA/metabolism , DNA/genetics , Nucleotide Motifs , Protein Binding , Cell Line, Tumor
2.
Breast Cancer Res Treat ; 192(1): 89-100, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35098437

ABSTRACT

PURPOSE: ITGA2 encodes the integrin, α2 which mediates metastatic progression, and is a predictor of poor prognosis and chemoresistance in breast cancer. Decreased ITGA2 promoter methylation is implicated as a driver of increased gene expression in aggressive prostate and pancreatic tumours, however the contribution of altered methylation to ITGA2 expression changes in breast tumours has not been examined. METHODS: ITGA2 gene methylation and gene expression was examined in publicly available breast cancer datasets, and ITGA2 promoter methylation was mapped by targeted bisulphite sequencing analysis in breast tumour cell lines. The expression of a putative regulatory long noncoding RNA (lncRNA) was examined by qPCR and its' functionality was investigated using gene knockdown (antisense oligonucleotides) and over expression in breast cancer cell lines. RESULTS: In breast tumours and breast cancer cell lines the ITGA2 promoter is largely unmethylated, with gene expression variable in tumour subtypes, irrespective of promoter methylation. A novel lncRNA (AC025180.1;ENSG00000249899), named herein I2ALR, was identified at the ITGA2 gene locus, and was variably expressed in breast tumours and breast cancer cell subtypes. I2LAR knockdown resulted in upregulation of ITGA2 gene expression, whilst over-expression of I2ALR resulted in downregulation of ITGA2 mRNA. Further, examination of two downstream targets of ITGA2 associated with breast tumor stemness and metastasis (CCND1 and ACLY), revealed concomitant gene expression changes in response to I2ALR modulation. CONCLUSION: I2ALR represents a novel regulatory molecule targeting ITGA2 expression in breast tumours; a finding of significant and topical interest to the development of therapeutics targeting this integrin.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Breast Neoplasms/genetics , Cell Line, Tumor , DNA Methylation , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrins , Male , RNA, Long Noncoding/genetics
3.
Exp Cell Res ; 392(2): 112055, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32376286

ABSTRACT

BACKGROUND: Examination of epigenetic changes at the ITGB4 gene promoter reveals altered methylation at different stages of prostate tumour progression and these changes may, in part, explain the complex patterns of gene expression of this integrin observed. Transcriptional re-programming perturbs expression of cell adhesion molecules and underpins metastatic tumour cell behaviour. Decreasing expression of the cell adhesion molecule ITGB4, which encodes the beta subunit of the integrin, alpha6 beta4 (α6ß4), has been correlated with increased tumour aggressiveness and metastasis in multiple tumour types including prostate cancer. Paradoxically, in vitro studies in tumour cell models demonstrate that ITGB4 mediates cell mobility and invasion. Herein we examined whether transcriptional re-programming by methylation influenced ITGB4 gene expression at different stages of prostate cancer progression. Bisulphite sequencing of a large CpG island in the ITGB4 gene promoter identified differentially methylated regions in prostate cancer cell lines representing a localised tumour (22Rv1), lymph node metastasis (LNCaP), and a bone metastasis (PC-3). The highest levels of methylation were observed in the CpG island surrounding the ITGB4 transcription start site in PC-3 cells, and this observation also correlated with higher gene expression of ITGB4 in these cells. Furthermore, PC-3 cells expressed two distinct transcripts, using an alternate transcription start site, which was not detected in other cell lines. In prostate tumour biopsy samples, patterns of methylation across the ITGB4 promoter were similar overall in matched primary and metastatic samples (n = 4 pairs), with a trend toward loss of methylation at specific sites in metastatic lesions.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Integrin beta4/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , CpG Islands , Humans , Integrin beta4/metabolism , Male , Tumor Cells, Cultured
4.
J Cell Physiol ; 233(4): 3439-3453, 2018 04.
Article in English | MEDLINE | ID: mdl-28926098

ABSTRACT

Integrins are transmembrane adhesion receptors that play an important role in hematopoiesis by facilitating interactions between hematopoietic cells and extracellular matrix components of the bone marrow and hematopoietic tissues. These interactions are important in regulating the function, proliferation, and differentiation of hematopoietic cells, as well as their homing and mobilization in the bone marrow. Not surprisingly altered expression and function of integrins plays a key role in the development and progression of cancer including leukemias. However, the regulation of integrin gene expression is not well characterized and the mechanisms by which integrin genes are disrupted in cancer remain unclear. Here we demonstrate for the first time that a key regulator of hematopoiesis, RUNX1, binds to and regulates the promoters of both the ITGA6 and ITGB4 genes in myeloid cells. The ITGA6 and ITGB4 integrin genes form the α6ß4 integrin receptor. However, our data indicate that RUNX1 functions differently at these two promoters. RUNX1 regulates ITGA6 through a consensus RUNX1 binding motif in its promoter. In contrast, although the ITGB4 promoter is also activated by RUNX1, it does so in the absence of a recognized consensus RUNX1 binding motif. Furthermore, our data suggest that regulation of ITGB4 may involve interactions between the promoter and upstream regulatory elements.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation, Developmental/genetics , Integrin alpha6/metabolism , Integrin beta4/metabolism , Myeloid Cells/metabolism , Cell Differentiation/genetics , Embryo, Mammalian/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mutation/genetics , Promoter Regions, Genetic/genetics
5.
J Cell Physiol ; 231(3): 688-97, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26241708

ABSTRACT

Fucoidan, a natural component of seaweeds, is reported to have immunomodulatory and anti-tumor effects. The mechanisms underpinning these activities remain poorly understood. In this study, the cytotoxicity and anti-tumor activities of fucoidan were investigated in acute myeloid leukemia (AML) cells. The human AML cell lines NB4, KG1a, HL60, and K562 were treated with fucoidan and cell cycle, cell proliferation, and expression of apoptotic pathways molecules were analyzed. Fucoidan suppressed the proliferation and induced apoptosis through the intrinsic and extrinsic pathways in the acute promyelocytic leukemia (APL) cell lines NB4 and HL60, but not in KG1a and K562 cells. In NB4 cells, apoptosis was caspase-dependent as it was significantly attenuated by pre-treatment with a pan-caspase inhibitor. P21/WAF1/CIP1 was significantly up-regulated leading to cell cycle arrest. Fucoidan decreased the activation of ERK1/2 and down-regulated the activation of AKT through hypo-phosphorylation of Thr(308) residue but not Ser(473). In vivo, a xenograft model using the NB4 cells was employed. Mice were fed with fucoidan and tumor growth was measured following inoculation with NB4 cells. Subsequently, splenic natural killer (NK) cell cytotoxic activity was also examined. Oral doses of fucoidan significantly delayed tumor growth in the xenograft model and increased cytolytic activity of NK cells. Taken together, these data suggest that the selective inhibitory effect of fucoidan on APL cells and its protective effect against APL development in mice warrant further investigation of fucoidan as a useful agent in treatment of certain types of leukemia.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Leukemia, Promyelocytic, Acute/pathology , Polysaccharides/pharmacology , Animals , Cell Line, Tumor , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Mice, Inbred BALB C
6.
J Cell Biochem ; 117(1): 49-58, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26060100

ABSTRACT

Activation of cytokine signaling via the leukemia inhibitory factor receptor (LIFR) plays an integral role in hematopoiesis, osteogenesis, and placental development, along with mediating neurotrophic mechanisms. However, the regulatory control of the LIFR gene has remained largely unexplored. Here, we characterize the LIFR gene as a novel target of the RUNX1 transcription factor. The RUNX1 transcription factor is an essential regulator of hematopoiesis and is a frequent target of point mutations and chromosomal alterations in leukemia. RUNX1 regulates hematopoiesis through its control of genes important for hematopoietic cell growth, proliferation, and differentiation, including a number of cytokines and cytokine receptors. LIFR is regulated by two alternate promoters: a placental-specific and a ubiquitously active general promoter. We show that both of these promoters are regulated by RUNX1. However, in myeloid cells LIFR expression is driven solely by the general LIFR promoter with our data indicating that the placental promoter is epigenetically silenced in these cells. While RUNX1 activates the LIFR general promoter, the oncogenic RUNX1-ETO fusion protein generated by the t(8;21) translocation commonly associated with acute myeloid leukemia represses promoter activity. The data presented here establish LIFR as a transcriptional target of RUNX1 and suggest that disruption of RUNX1 activity in myeloid cells may result in altered LIFR signaling in these cells.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Receptors, OSM-LIF/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Chromatin Immunoprecipitation , Chromosome Aberrations , Core Binding Factor Alpha 2 Subunit/genetics , Humans , Myeloid Cells/metabolism , Point Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Binding/physiology , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, OSM-LIF/genetics
7.
Prostate ; 75(7): 723-34, 2015 May.
Article in English | MEDLINE | ID: mdl-25662931

ABSTRACT

BACKGROUND: Integrin alpha2 beta1 (α2 ß1 ) plays an integral role in tumour cell invasion, metastasis and angiogenesis, and altered expression of the receptor has been linked to tumour prognosis in several solid tumours. However, the relationship is complex, with both increased and decreased expression associated with different stages of tumour metastases in several tumour types. The ITGA2 gene, which codes for the α2 subunit, was examined to investigate whether a large CpG island associated with its promoter region is involved in the differential expression of ITGA2 observed in prostate cancer. METHODS: Bisulphite sequencing of the ITGA2 promoter was used to assess methylation in formalin-fixed paraffin-embedded (FFPE) prostate tumour specimens and prostate cancer cell lines, PC3, 22Rv1 and LNCaP. Changes in ITGA2 mRNA expression were measured using quantitative PCR. ITGA2 functionality was interrogated using cell migration scratch assays and siRNA knockdown experiments. RESULTS: Bisulphite sequencing revealed strikingly decreased methylation at key CpG sites within the promoter of tumour samples, when compared with normal prostate tissue. Altered methylation of this CpG island is also associated with differences in expression in the non-invasive LNCaP, and the highly metastatic PC3 and 22Rv1 prostate cancer cell lines. Further bisulphite sequencing confirmed that selected CpGs were highly methylated in LNCaP cells, whilst only low levels of methylation were observed in PC3 and 22Rv1 cells, correlating with ITGA2 transcript levels. Examination of the increased expression of ITGA2 was shown to influence migratory potential via scratch assay in PC3, 22Rv1 and LNCaP cells, and was confirmed by siRNA knockdown experiments. CONCLUSIONS: Taken together, our data supports the assertion that epigenetic modification of the ITGA2 promoter is a mechanism by which ITGA2 expression is regulated.


Subject(s)
Integrin alpha5beta1/genetics , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha5beta1/biosynthesis , Male , Middle Aged , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
8.
Mol Med ; 21: 400-9, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25822796

ABSTRACT

The field of epigenetics and our understanding of the mechanisms that regulate the establishment, maintenance and heritability of epigenetic patterns continue to grow at a remarkable rate. This information is providing increased understanding of the role of epigenetic changes in disease, insight into the underlying causes of these epigenetic changes and revealing new avenues for therapeutic intervention. Epigenetic modifiers are increasingly being pursued as therapeutic targets in a range of diseases, with a number of agents targeting epigenetic modifications already proving effective in diseases such as cancer. Although it is well established that DNA mutations and aberrant expression of epigenetic modifiers play a key role in disease, attention is now turning to the interplay between genetic and epigenetic factors in complex disease etiology. The role of genetic variability in determining epigenetic profiles, which can then be modified by environmental and stochastic factors, is becoming more apparent. Understanding the interplay between genetic and epigenetic factors is likely to aid in identifying individuals most likely to benefit from epigenetic therapies. This goal is coming closer to realization because of continual advances in laboratory and statistical tools enabling improvements in the integration of genomic, epigenomic and phenotypic data.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic , RNA, Untranslated/genetics , Euchromatin/genetics , Genetic Therapy , Genome, Human , Heterochromatin/genetics , Histones/genetics , Humans , Mutation
9.
Mar Drugs ; 13(4): 2327-46, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25874926

ABSTRACT

There is a wide variety of cancer types yet, all share some common cellular and molecular behaviors. Most of the chemotherapeutic agents used in cancer treatment are designed to target common deregulated mechanisms within cancer cells. Many healthy tissues are also affected by the cytotoxic effects of these chemical agents. Fucoidan, a natural component of brown seaweed, has anti-cancer activity against various cancer types by targeting key apoptotic molecules. It also has beneficial effects as it can protect against toxicity associated with chemotherapeutic agents and radiation. Thus the synergistic effect of fucoidan with current anti-cancer agents is of considerable interest. This review discusses the mechanisms by which fucoidan retards tumor development, eradicates tumor cells and synergizes with anti-cancer chemotherapeutic agents. Challenges to the development of fucoidan as an anti-cancer agent will also be discussed.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drugs, Investigational/pharmacology , Models, Biological , Neoplasms/drug therapy , Polysaccharides/pharmacology , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Drug Evaluation, Preclinical , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Drugs, Investigational/therapeutic use , Functional Food/analysis , Humans , MAP Kinase Signaling System/drug effects , Neoplasm Metastasis/prevention & control , Neoplasms/metabolism , Neoplasms/pathology , Phaeophyceae/chemistry , Polysaccharides/administration & dosage , Polysaccharides/adverse effects , Polysaccharides/therapeutic use , Seaweed/chemistry
10.
BMC Cancer ; 14: 808, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25369795

ABSTRACT

BACKGROUND: Radiotherapy is a chosen treatment option for prostate cancer patients and while some tumours respond well, up to 50% of patients may experience tumour recurrence. Identification of functionally relevant predictive biomarkers for radioresponse in prostate cancer would enable radioresistant patients to be directed to more appropriate treatment options, avoiding the side-effects of radiotherapy. METHODS: Using an in vitro model to screen for novel biomarkers of radioresistance, transcriptome analysis of a radioresistant (PC-3) and radiosensitive (LNCaP) prostate cancer cell line was performed. Following pathway analysis candidate genes were validated using qRT-PCR. The DNA repair pathway in radioresistant PC-3 cells was then targeted for radiation sensitization using the PARP inhibitor, niacinimide. RESULTS: Opposing regulation of a DNA repair and replication pathway was observed between PC-3 and LNCaP cells from RNA-seq analysis. Candidate genes BRCA1, RAD51, FANCG, MCM7, CDC6 and ORC1 were identified as being significantly differentially regulated post-irradiation. qRT-PCR validation confirmed BRCA1, RAD51 and FANCG as being significantly differentially regulated at 24 hours post radiotherapy (p-value =0.003, 0.045 and 0.003 respectively). While the radiosensitive LNCaP cells down-regulated BRCA1, FANCG and RAD51, the radioresistant PC-3 cell line up-regulated these candidates to promote cell survival post-radiotherapy and a similar trend was observed for MCM7, CDC6 and ORC1. Inhibition of DNA repair using niacinamide sensitised the radioresistant cells to irradiation, reducing cell survival at 2 Gy from 66% to 44.3% (p-value =0.02). CONCLUSIONS: These findings suggest that the DNA repair candidates identified via RNA-seq hold potential as both targets for radiation sensitization and predictive biomarkers in prostate cancer.


Subject(s)
Biomarkers, Tumor/genetics , DNA Repair/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/genetics , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Repair/drug effects , Down-Regulation/radiation effects , Enzyme Inhibitors/pharmacology , Fanconi Anemia Complementation Group G Protein/genetics , Gene Expression Profiling , Humans , Male , Minichromosome Maintenance Complex Component 7/genetics , Niacinamide/pharmacology , Nuclear Proteins/genetics , Origin Recognition Complex/genetics , Poly(ADP-ribose) Polymerase Inhibitors , RNA, Messenger/analysis , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Radiation-Sensitizing Agents/therapeutic use , Up-Regulation/radiation effects
11.
Mult Scler ; 20(2): 135-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24493701

ABSTRACT

It is now well established that both genetic and environmental factors contribute to and interact in the development of multiple sclerosis (MS). However, the currently described causal genetic variants do not explain the majority of the heritability of MS, resulting in 'missing heritability'. Epigenetic mechanisms, which principally include DNA methylation, histone modifications and microRNA-mediated post-transcriptional gene silencing, may contribute a significant component of this missing heritability. As the development of MS is a dynamic process potentially starting with inflammation, then demyelination, remyelination and neurodegeneration, we have reviewed the dynamic epigenetic changes in these aspects of MS pathogenesis and describe how environmental risk factors may interact with epigenetic changes to manifest in disease.


Subject(s)
Epigenesis, Genetic/physiology , Genetic Predisposition to Disease/genetics , Multiple Sclerosis/genetics , Humans
12.
Immunology ; 139(3): 285-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23521628

ABSTRACT

T cells are exquisitely poised to respond rapidly to pathogens and have proved an instructive model for exploring the regulation of inducible genes. Individual genes respond to antigenic stimulation in different ways, and it has become clear that the interplay between transcription factors and the chromatin platform of individual genes governs these responses. Our understanding of the complexity of the chromatin platform and the epigenetic mechanisms that contribute to transcriptional control has expanded dramatically in recent years. These mechanisms include the presence/absence of histone modification marks, which form an epigenetic signature to mark active or inactive genes. These signatures are dynamically added or removed by epigenetic enzymes, comprising an array of histone-modifying enzymes, including the more recently recognized chromatin-associated signalling kinases. In addition, chromatin-remodelling complexes physically alter the chromatin structure to regulate chromatin accessibility to transcriptional regulatory factors. The advent of genome-wide technologies has enabled characterization of the chromatin landscape of T cells in terms of histone occupancy, histone modification patterns and transcription factor association with specific genomic regulatory regions, generating a picture of the T-cell epigenome. Here, we discuss the multi-layered regulation of inducible gene expression in the immune system, focusing on the interplay between transcription factors, and the T-cell epigenome, including the role played by chromatin remodellers and epigenetic enzymes. We will also use IL2, a key inducible cytokine gene in T cells, as an example of how the different layers of epigenetic mechanisms regulate immune responsive genes during T-cell activation.


Subject(s)
Chromatin/metabolism , Epigenomics , Gene Expression Regulation , T-Lymphocytes/metabolism , Humans , Lymphocyte Activation , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Cell Oncol (Dordr) ; 46(2): 239-250, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36512308

ABSTRACT

BACKGROUND: Integrins are integral to cell signalling and management of the extracellular matrix, and exquisite regulation of their expression is essential for a variety of cell signalling pathways, whilst disordered regulation is a key driver of tumour progression and metastasis. Most recently non-coding RNAs in the form of micro-RNA (miRNA) and long non-coding RNA (lncRNA) have emerged as a key mechanism by which tissue dependent gene expression is controlled. Whilst historically these molecules have been poorly understood, advances in 'omic' technologies and a greater understanding of non-coding regions of the genome have revealed that non-coding RNAs make up a large proportion of the transcriptome. CONCLUSIONS AND PERSPECTIVES: This review examines the regulation of integrin genes by ncRNAs, provides and overview of their mechanism of action and highlights how exploitation of these discoveries is informing the development of novel chemotherapeutic agents in the treatment of cancer. MiRNA molecules have been the most extensively characterised and negatively regulate most integrin genes, classically regulating genes through binding to recognition sequences in the mRNA 3'-untranslated regions of gene transcripts. LncRNA mechanisms of action are now being elucidated and appear to be more varied and complex, and may counter miRNA molecules, directly engage integrin mRNA transcripts, and guide or block both transcription factors and epigenetic machinery at integrin promoters or at other points in integrin regulation. Integrins as therapeutic targets are of enormous interest given their roles as oncogenes in a variety of tumours, and emerging therapeutics mimicking ncRNA mechanisms of action are already being trialled.


Subject(s)
MicroRNAs , Neoplasms , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Untranslated/genetics , Neoplasms/genetics , MicroRNAs/genetics , RNA, Messenger
14.
Mol Cell Neurosci ; 46(1): 213-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20840869

ABSTRACT

Nuclear factor kappaB (NFκB) is a key transcriptional regulator of inflammatory genes. We investigated the modulatory effects of olfactory ensheathing cells (OECs), microglia and meningeal fibroblasts on translocation of NFκB to astrocyte nuclei. The percentage of activated astrocytes in co-cultures with OECs was significantly less than for co-cultures with microglia (p<0.001) and fibroblasts (p<0.05). Phorbol myristate acetate (PMA) and calcium ionophore stimulation of p65 NFκB translocation to nuclei provided an in vitro model of astrocyte inflammatory activation. Soluble factors released by OECs significantly moderated the astrocytic NFκB translocation induced by either PMA/calcium ionophore or microglia-derived factors (p<0.001). Insulin-like growth factor-1 may contribute to these effects, since it is expressed by OECs and also significantly moderated the astrocytic NFκB translocation (p<0.05), albeit insufficiently to fully account for the OEC-induced moderation (p<0.01). Olfactory ensheathing cells significantly moderated the increased transcription of the pro-inflammatory cytokine, granulocyte macrophage-colony stimulating factor in the activated astrocytes (p<0.01). These results suggest that transplanted OECs could improve neural repair after CNS injury by moderating astrocyte activation.


Subject(s)
Astrocytes/metabolism , NF-kappa B/metabolism , Schwann Cells/physiology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Calcium/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Coculture Techniques , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Insulin-Like Growth Factor I/metabolism , Ionophores/pharmacology , Meninges/cytology , Microglia/cytology , Microglia/drug effects , Microglia/physiology , Protein Transport/physiology , Rats , Rats, Wistar , Schwann Cells/cytology , Tetradecanoylphorbol Acetate/pharmacology
15.
J Exp Med ; 197(4): 413-23, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12591900

ABSTRACT

Granulocyte/macrophage colony-stimulating factor (GM-CSF) is a key cytokine in myelopoiesis and aberrant expression is associated with chronic inflammatory disease and myeloid leukemias. This aberrant expression is often associated with constitutive nuclear factor (NF)-kappaB activation. To investigate the relationship between NF-kappaB and GM-CSF transcription in a chromatin context, we analyzed the chromatin structure of the GM-CSF gene in T cells and the role of NF-kappaB proteins in chromatin remodeling. We show here that chromatin remodeling occurs across a region of the GM-CSF gene between -174 and +24 upon T cell activation, suggesting that remodeling is limited to a single nucleosome encompassing the proximal promoter. Nuclear NF-kappaB levels appear to play a critical role in this process. In addition, using an immobilized template assay we found that the ATPase component of the SWI/SNF chromatin remodeling complex, brg1, is recruited to the GM-CSF proximal promoter in an NF-kappaB-dependent manner in vitro. These results suggest that chromatin remodeling across the GM-CSF promoter in T cells is a result of recruitment of SWI/SNF type remodeling complexes by NF-kappaB proteins binding to the CD28 response region of the promoter.


Subject(s)
Chromatin/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , T-Lymphocytes/immunology , CD28 Antigens , DNA Helicases , Humans , Jurkat Cells , Nuclear Proteins/physiology , Nucleosomes/metabolism , RNA, Messenger/analysis , Response Elements , Transcription Factors/physiology
16.
Epigenetics ; 14(10): 989-1002, 2019 10.
Article in English | MEDLINE | ID: mdl-31208284

ABSTRACT

Many cancer therapies operate by inducing double-strand breaks (DSBs) in cancer cells, however treatment-resistant cells rapidly initiate mechanisms to repair damage enabling survival. While the DNA repair mechanisms responsible for cancer cell survival following DNA damaging treatments are becoming better understood, less is known about the role of the epigenome in this process. Using prostate cancer cell lines with differing sensitivities to radiation treatment, we analysed the DNA methylation profiles prior to and following a single dose of radiotherapy (RT) using the Illumina Infinium HumanMethylation450 BeadChip platform. DSB formation and repair, in the absence and presence of the DNA hypomethylating agent, 5-azacytidine (5-AzaC), were also investigated using γH2A.X immunofluorescence staining. Here we demonstrate that DNA methylation is generally stable following a single dose of RT; however, a small number of CpG sites are stably altered up to 14 d following exposure. While the radioresistant and radiosensitive cells displayed distinct basal DNA methylation profiles, their susceptibility to DNA damage appeared similar demonstrating that basal DNA methylation has a limited influence on DSB induction at the regions examined. Recovery from DSB induction was also similar between these cells. Treatment with 5-AzaC did not sensitize resistant cells to DNA damage, but rather delayed recruitment of phosphorylated BRCA1 (S1423) and repair of DSBs. These results highlight that stable epigenetic changes are possible following a single dose of RT and may have significant clinical implications for cancer treatment involving recurrent or fractionated dosing regimens.


Subject(s)
Azacitidine/pharmacology , DNA Damage , DNA Methylation , Prostatic Neoplasms/genetics , BRCA1 Protein/metabolism , Cell Line, Tumor , CpG Islands/drug effects , CpG Islands/radiation effects , DNA Methylation/drug effects , DNA Methylation/radiation effects , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/radiation effects , High-Throughput Nucleotide Sequencing , Humans , Male , PC-3 Cells , Phosphorylation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Radiation Tolerance , Sequence Analysis, DNA
17.
Pathology ; 50(6): 622-628, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30149991

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN 1) has marked severity variation between individuals with the same mutation. To investigate any relationship between promoter methylation and clinical features, blood and tissue samples were collected from 16 members of the Tasman 1 MEN 1 kindred carrying a common splice site mutation and 7 patients with sporadic MEN 1. Methylation at 39 CpGs in the MEN1 promoter were assessed in formalin fixed, paraffin embedded parathyroid tissue. Clinical disease severity markers included age at first parathyroid operation, parathyroid hormone level and corrected serum calcium levels. Six patients with sporadic hyperparathyroidism were used for comparison. Minimal methylation was observed in all patients across CpG sites 1-23. In contrast, hypermethylation was observed at CpG sites 24-31 in MEN 1 patients, a pattern not observed in patients with non-MEN 1 parathyroid disease. Mean methylation at sites 24-31 was significantly correlated with age at first parathyroid operation (r = 0.652, p = 0.041). A permutation test, utilising the mean correlation coefficient (r = -0.401) revealed a possible association between relative PHPT severity and methylation score for each significant CpG site (p < 0.103). This novel study reveals evidence supporting a possible association between altered MEN1 promoter methylation and clinical severity of disease.


Subject(s)
DNA Methylation/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Adult , Female , Humans , Hyperparathyroidism/genetics , Male , Middle Aged , Promoter Regions, Genetic/genetics , Young Adult
18.
Nucleic Acids Res ; 33(1): 225-34, 2005.
Article in English | MEDLINE | ID: mdl-15647505

ABSTRACT

Granulocyte-macrophage colony stimulating factor (GM-CSF) plays a key role in myeloid cell function and is rapidly and transiently expressed in T cells in response to immune or inflammatory stimuli. Induction of GM-CSF gene expression is accompanied by changes in chromatin structure across the proximal promoter region of the gene. We show that the promoter remodelling and subsequent gene transcription occurs with distinct signal and transcription factor requirements. Activation of the protein kinase C (PKC) signalling pathway is sufficient to induce changes in chromatin structure across the promoter, but both the PKC and calcium signalling pathways are required for efficient gene transcription. Although NFAT transcription factors contribute to GM-CSF gene transcription, they are not required for promoter remodelling. However, the presence of the nuclear factor-kappaB transcription factor, c-Rel, in the nucleus is strongly correlated with and required for the events of chromatin remodelling.


Subject(s)
Chromatin Assembly and Disassembly , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/physiology , Transcription, Genetic , Animals , Cell Nucleus/chemistry , Mice , NF-kappa B/analysis , Protein Biosynthesis , Proto-Oncogene Proteins c-rel/physiology
19.
Oncotarget ; 7(29): 46028-46041, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27329592

ABSTRACT

The morbidity and mortality associated with current therapies for acute promyelocytic leukemia (APL) remain a significant clinical concern, despite improvements in patient survival. Consequently, the development of adjuvant therapies that increase efficacy while reducing morbidities is important. Reducing the concentration of the toxic drugs in adjuvant therapy has the potential to reduce unwanted side effects. Therefore, this study aimed to determine the synergistic effects of fucoidan, an anti-tumor agent, with current APL therapies.When the human APL cell line, NB4, was treated in vitro with fucoidan plus ATO and ATRA at therapeutic and sub-therapeutic doses, there was an increase in sub-G0/G1 cells, annexin V/PI-positive-apoptotic cells and DNA fragmentation. This reduction in proliferation and increase in apoptosis was accompanied by enhanced myeloid differentiation as indicated by an increased expression of CD11b. This was not observed with the AML cell line Kasumi-1, suggesting specificity for APL.In vivo treatment of APL-bearing mice with fucoidan+ATRA or fucoidan+ATO delayed tumor growth, induced differentiation and increased tumor volume doubling time. The differentiated APL cells derived from the excised tumor mass exhibited decreased CD44 expression in fucoidan+ATRA treated mice. This could translate to decreased cell migration in APL patients.Our findings provide evidence supporting the use of fucoidan as an adjuvant therapeutic agent in the treatment of APL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Promyelocytic, Acute , Animals , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/administration & dosage , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oxides/administration & dosage , Polysaccharides/administration & dosage , Tretinoin/administration & dosage , Xenograft Model Antitumor Assays
20.
Clin Epigenetics ; 8: 75, 2016.
Article in English | MEDLINE | ID: mdl-27429663

ABSTRACT

BACKGROUND: Human methylome mapping in health and disease states has largely relied on Illumina Human Methylation 450k array (450k array) technology. Accompanying this has been the necessary evolution of analysis pipelines to facilitate data processing. The majority of these pipelines, however, cater for experimental designs where matched 'controls' or 'normal' samples are available. Experimental designs where no appropriate 'reference' exists remain challenging. Herein, we use data generated from our study of the inheritance of methylome profiles in families to evaluate the performance of eight normalisation pre-processing methods. Fifty individual samples representing four families were interrogated on five 450k array BeadChips. Eight normalisation methods were tested using qualitative and quantitative metrics, to assess efficacy and suitability. RESULTS: Stratified quantile normalisation combined with ComBat were consistently found to be the most appropriate when assessed using density, MDS and cluster plots. This was supported quantitatively by ANOVA on the first principal component where the effect of batch dropped from p < 0.01 to p = 0.97 after stratified QN and ComBat. Median absolute differences between replicated samples were the lowest after stratified QN and ComBat as were the standard error measures on known imprinted regions. Biological information was preserved after normalisation as indicated by the maintenance of a significant association between a known mQTL and methylation (p = 1.05e-05). CONCLUSIONS: A strategy combining stratified QN with ComBat is appropriate for use in the analyses when no reference sample is available but preservation of biological variation is paramount. There is great potential for use of 450k array data to further our understanding of the methylome in a variety of similar settings. Such advances will be reliant on the determination of appropriate methodologies for processing these data such as established here.


Subject(s)
DNA Methylation , Genome, Human , Oligonucleotide Array Sequence Analysis/standards , Sequence Analysis, DNA/standards , Adult , Aged , Aged, 80 and over , CpG Islands , Databases, Genetic , Female , Heredity , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Quantitative Trait Loci , Sequence Analysis, DNA/methods , Software , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL