Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cells ; 12(17)2023 08 29.
Article in English | MEDLINE | ID: mdl-37681897

ABSTRACT

Podocyte cellular injury and detachment from glomerular capillaries constitute a critical factor contributing to kidney disease. Notably, transcription factors are instrumental in maintaining podocyte differentiation and homeostasis. This study explores the hitherto uninvestigated expression of Nuclear Factor Erythroid 2-related Factor 1 (NFE2L1) in podocytes. We evaluated the podocyte expression of NFE2L1, Nuclear Factor Erythroid 2-related Factor 2 (NFE2L2), and NAD(P)H:quinone Oxidoreductase (NQO1) in 127 human glomerular disease biopsies using multiplexed immunofluorescence and image analysis. We found that both NFE2L1 and NQO1 expressions were significantly diminished across all observed renal diseases. Furthermore, we exposed human immortalized podocytes and ex vivo kidney slices to Puromycin Aminonucleoside (PAN) and characterized the NFE2L1 protein isoform expression. PAN treatment led to a reduction in the nuclear expression of NFE2L1 in ex vivo kidney slices and podocytes.


Subject(s)
Kidney Diseases , Podocytes , Humans , Gene Expression Regulation , Kidney , Kidney Glomerulus , NAD , NF-E2-Related Factor 1
2.
Front Physiol ; 12: 625762, 2021.
Article in English | MEDLINE | ID: mdl-34335284

ABSTRACT

Podocyte loss plays a pivotal role in the pathogenesis of glomerular disease. However, the mechanisms underlying podocyte damage and loss remain poorly understood. Although detachment of viable cells has been documented in experimental Diabetic Nephropathy, correlations between reduced podocyte density and disease severity have not yet been established. YAP, a mechanosensing protein, has recently been shown to correlate with glomerular disease progression, however, the underlying mechanism has yet to be fully elucidated. In this study, we sought to document podocyte density in Diabetic Nephropathy using an amended podometric methodology, and to investigate the interplay between YAP and cytoskeletal integrity during podocyte injury. Podocyte density was quantified using TLE4 and GLEPP1 multiplexed immunofluorescence. Fourteen Diabetic Nephropathy cases were analyzed for both podocyte density and cytoplasmic translocation of YAP via automated image analysis. We demonstrate a significant decrease in podocyte density in Grade III/IV cases (124.5 per 106 µm3) relative to Grade I/II cases (226 per 106 µm3) (Student's t-test, p < 0.001), and further show that YAP translocation precedes cytoskeletal rearrangement following injury. Based on these findings we hypothesize that a significant decrease in podocyte density in late grade Diabetic Nephropathy may be explained by early cytoplasmic translocation of YAP.

3.
Clin Sci (Lond) ; 134(11): 1245-1253, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32501496

ABSTRACT

Chronic kidney disease (CKD) substantially reduces quality of life and leads to premature death for thousands of people each year. Dialysis and kidney organ transplants remain prevalent therapeutic avenues but carry significant medical, economic and social burden. Podocytes are responsible for blood filtration selectivity in the kidney, where they extend a network of foot processes (FPs) from their cell bodies which surround endothelial cells and interdigitate with those on neighbouring podocytes to form narrow slit diaphragms (SDs). During aging, some podocytes are lost naturally but accelerated podocyte loss is a hallmark of CKD. Insights into the origin of degenerative podocyte loss will help answer important questions about kidney function and lead to substantial health benefits. Here, approaches that uncover insights into podocyte mechanobiology are reviewed, both those that interrogate the biophysical properties of podocytes and how the external physical environment affects podocyte behaviour, and also those that interrogate the biophysical effects that podocytes exert on their surroundings.


Subject(s)
Biophysics , Health , Kidney Diseases/pathology , Podocytes/pathology , Animals , Biomechanical Phenomena , Humans , Stress, Mechanical
4.
Sci Rep ; 9(1): 7643, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31113993

ABSTRACT

Gemcitabine is a fluoropyrimidine analogue that is used as a mainstay of chemotherapy treatment for pancreatic and ovarian cancers, amongst others. Despite its widespread use, gemcitabine achieves responses in less than 10% of patients with metastatic pancreatic cancer and has a very limited impact on overall survival due to intrinsic and acquired resistance. NUC-1031 (Acelarin), a phosphoramidate transformation of gemcitabine, was the first anti-cancer ProTide to enter the clinic. We find it displays important in vitro cytotoxicity differences to gemcitabine, and a genome-wide CRISPR/Cas9 genetic screening approach identified only the pyrimidine metabolism pathway as modifying cancer cell sensitivity to NUC-1031. Low deoxycytidine kinase expression in tumour biopsies from patients treated with gemcitabine, assessed by immunostaining and image analysis, correlates with a poor prognosis, but there is no such correlation in tumour biopsies from a Phase I cohort treated with NUC-1031.


Subject(s)
Antineoplastic Agents/toxicity , Biomarkers, Tumor/genetics , Cytidine Monophosphate/analogs & derivatives , Deoxycytidine Kinase/genetics , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/genetics , Pancreatic Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , CRISPR-Cas Systems , Clinical Trials, Phase I as Topic , Cytidine Monophosphate/therapeutic use , Cytidine Monophosphate/toxicity , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Deoxycytidine Kinase/metabolism , Female , HEK293 Cells , Humans , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Gemcitabine
5.
Cancer Drug Resist ; 2(2): 356-364, 2019.
Article in English | MEDLINE | ID: mdl-35582719

ABSTRACT

Receptor tyrosine kinase inhibitors have been a standard first-line therapy for renal cell carcinoma (RCC) for over a decade. Although they stabilize the disease, they are unable to remove all tumor cells, leading to relapse. Moreover, both intrinsic and acquired resistance to therapy are a significant health burden. In order to overcome resistance, several combination therapies have been recently approved by the FDA. Another approach takes advantage of altered metabolism in tumor cells, which switch to alternative metabolic pathways to sustain their rapid growth and proliferation. CB-839 is a small molecule inhibitor of kidney type glutaminase (GLS). GLS is often upregulated in glutamine addicted cancers, enhancing glutamine metabolism for the production of energy and the biosynthesis of various cellular building blocks. CB-839 is currently in clinical trials for several tumors, including clear cell (cc)RCC, both as monotherapy and in combination with the approved therapeutic agents everolimus, cabozantinib and nivolumab. Early results of Phase 1/2 clinical trials look promising, especially for CB-839 plus cabozantinib, and all combinations seem to be well tolerated. However, cancer cells can activate compensatory pathways to overcome glutaminolysis inhibition. Therefore, genetic and metabolomic studies are crucial for the successful implementation of CB-839 alone or in combination in subgroups of ccRCC patients.

6.
Sci Adv ; 4(6): eaap8030, 2018 06.
Article in English | MEDLINE | ID: mdl-29963620

ABSTRACT

In the healthy kidney, specialized cells called podocytes form a sophisticated blood filtration apparatus that allows excretion of wastes and excess fluid from the blood while preventing loss of proteins such as albumin. To operate effectively, this filter is under substantial hydrostatic mechanical pressure. Given their function, it is expected that the ability to apply mechanical force is crucial to the survival of podocytes. However, to date, podocyte mechanobiology remains poorly understood, largely because of a lack of experimental data on the forces involved. We perform quantitative, continuous, nondisruptive, and high-resolution measurements of the forces exerted by differentiated podocytes in real time using a recently introduced functional imaging modality for continuous force mapping. Using an accepted model for podocyte injury, we find that injured podocytes experience near-complete loss of cellular force transmission but that this loss of force is reversible under certain conditions. The observed changes in force correlate with F-actin rearrangement and reduced expression of podocyte-specific proteins. By introducing robust and high-throughput mechanical phenotyping and by demonstrating the significance of mechanical forces in podocyte injury, this research paves the way to a new level of understanding of the kidney. In addition, in an advance over established force mapping techniques, we integrate cellular force measurements with immunofluorescence and perform continuous long-term force measurements of a cell population. Hence, our approach has general applicability to a wide range of biomedical questions involving mechanical forces.


Subject(s)
Biomarkers , Biomechanical Phenomena , Mechanotransduction, Cellular , Podocytes/metabolism , Animals , Cell Differentiation , Cytoskeleton/metabolism , Fluorescent Antibody Technique , Humans , Mice , Phenotype , Podocytes/cytology , Stress, Physiological
7.
Genes (Basel) ; 7(7)2016 Jul 18.
Article in English | MEDLINE | ID: mdl-27438856

ABSTRACT

Genome sequencing is now a common procedure, but prior to this, screening experiments using protein baits was one of the routinely used methods that, occasionally, allowed the identification of new gene products. One such experiment uncovered the gene product called willin/human Expanded/FRMD6. Initial characterization studies found that willin bound phospholipids and was strongly co-localised with actin. However, subsequently, willin was found to be the closest human sequence homologue of the Drosophila protein Expanded (Ex), sharing 60% homology with the Ex FERM domain. This in turn suggested, and then was proven that willin could activate the Hippo signalling pathway. This review describes the increasing body of knowledge about the actions of willin in a number of cellular functions related to cancer. However, like many gene products involved in aspects of cell signalling, a convincing direct role for willin in cancer remains tantalisingly elusive, at present.

8.
J Mol Cell Biol ; 8(5): 439-455, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27190314

ABSTRACT

Crumbs 3 (CRB3) is a component of epithelial junctions, which has been implicated in apical-basal polarity, apical identity, apical stability, cell adhesion, and cell growth. CRB3 undergoes alternative splicing to yield two variants: CRB3a and CRB3b. Here, we describe novel data demonstrating that, as with previous studies on CRB3a, CRB3b also promotes the formation of tight junctions (TJs). However, significantly we demonstrate that the 4.1-ezrin-radixin-moesin-binding motif of CRB3b is required for CRB3b functionality and that ezrin binds to the FBM of CRB3b. Furthermore, we show that ezrin contributes to CRB3b functionality and the correct distribution of TJ proteins. We demonstrate that both CRB3 isoforms are required for the production of functionally mature TJs and also the localization of ezrin to the plasma membrane. Finally, we demonstrate that reduced CRB3b expression in head and neck squamous cell carcinoma (HNSCC) correlates with cytoplasmic ezrin, a biomarker for aggressive disease, and shows evidence that while CRB3a expression has no effect, low CRB3b and high cytoplasmic ezrin expression combined may be prognostic for HNSCC.

9.
Acad Pathol ; 2(4): 2374289515616972, 2015.
Article in English | MEDLINE | ID: mdl-28725753

ABSTRACT

We retrospectively studied the expression of Yes-associated protein (YAP) using immunohistochemical staining in 10 cases of head and neck squamous cell carcinoma with associated perineural invasion. We find that fibroblasts in areas associated with perineural invasion show higher levels of nuclear YAP compared to fibroblasts in the stroma of normal mucosa, with a median cell count of 35.4 per high-power field in the former and 3.9 in the latter. No differences were observed between the expression of YAP phosphorylated at Ser127 in the tumoral stroma compared to that in the normal mucosa, with a median cell count expression of 4.9 in the former versus 5.0 in the latter. Therefore, a strong and increased nuclear YAP expression in fibroblasts associated with perineural invasion in head and neck squamous cell carcinoma suggests that YAP-mediated transcription programs in these fibroblasts may contribute to perineural invasion.

10.
FEBS J ; 280(23): 5949-56, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24118991

ABSTRACT

Histopathology, the examination of an architecturally artefactual, two-dimensional and static image remains a potent tool allowing diagnosis and empirical expectation of prognosis. Considerable optimism exists that the advent of molecular genetic testing and other biomarker strategies will improve or even replace this ancient technology. A number of biomarkers already add considerable value for prediction of whether a treatment will work. In this short review we argue that a systems medicine approach to pathology will not seek to replace traditional pathology, but rather augment it. Systems approaches need to incorporate quantitative morphological, protein, mRNA and DNA data. A significant challenge for clinical implementation of systems pathology is how to optimize information available from tissue, which is frequently sub-optimal in quality and amount, and yet generate useful predictive models that work. The transition of histopathology to systems pathophysiology and the use of multiscale data sets usher in a new era in diagnosis, prognosis and prediction based on the analysis of human tissue.


Subject(s)
Biomarkers, Tumor/metabolism , Molecular Diagnostic Techniques , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , Systems Biology , Humans , Neoplasms/metabolism , Precision Medicine , Prognosis
11.
Biochem J ; 452(2): 183-93, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23662806

ABSTRACT

Our understanding of the FERM (4.1/ezrin/radixin/moesin) protein family has been rapidly expanding in the last few years, with the result that many new physiological functions have been ascribed to these biochemically unique proteins. In the present review, we will discuss a number of new FRMD (FERM domain)-containing proteins that were initially discovered from genome sequencing but are now being established through biochemical and genetic studies to be involved both in normal cellular processes, but are also associated with a variety of human diseases.


Subject(s)
Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/physiology , Cytoskeletal Proteins/genetics , Forecasting , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/physiology , Multigene Family/genetics , Protein Structure, Tertiary
12.
PLoS One ; 8(4): e60028, 2013.
Article in English | MEDLINE | ID: mdl-23593160

ABSTRACT

Willin/FRMD6 was first identified in the rat sciatic nerve, which is composed of neurons, Schwann cells, and fibroblasts. Willin is an upstream component of the Hippo signaling pathway, which results in the inactivation of the transcriptional co-activator YAP through Ser127 phosphorylation. This in turn suppresses the expression of genes involved in cell growth, proliferation and cancer development ensuring the control of organ size, cell contact inhibition and apoptosis. Here we show that in the mammalian sciatic nerve, Willin is predominantly expressed in fibroblasts and that Willin expression activates the Hippo signaling cascade and induces YAP translocation from the nucleus to the cytoplasm. In addition within these cells, although it inhibits cellular proliferation, Willin expression induces a quicker directional migration towards scratch closure and an increased expression of factors linked to nerve regeneration. These results show that Willin modulates sciatic nerve fibroblast activity indicating that Willin may have a potential role in the regeneration of the peripheral nervous system.


Subject(s)
Fibroblasts/cytology , Fibroblasts/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Sciatic Nerve/cytology , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Movement , Cell Proliferation , Ephrin-B2/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation , Hippo Signaling Pathway , Homeostasis , Intracellular Signaling Peptides and Proteins/genetics , Mice , Phosphoproteins/metabolism , YAP-Signaling Proteins
13.
Head Neck Pathol ; 6(2): 232-43, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22228071

ABSTRACT

Members of the 4.1 superfamily of proteins, including ezrin, moesin, merlin, and willin regulate many normal physiologic processes such as cellular shape, motility, and proliferation. In addition, they contribute both to tumor development and tumor progression. We reported previously that strong cytoplasmic ezrin expression was independently associated with poorer patient survival. One hundred and thirty-one histologically confirmed primary head and neck squamous cell carcinomas were examined prospectively for cancer progression and survival at a large health care center in the Bronx, NY, USA. Immunohistochemical analysis of ezrin, moesin, merlin, and willin expression in tissue microarray samples of primary head and neck squamous cell carcinoma revealed a significant association of increased cytoplasmic ezrin with poor cancer survival. Global RNA analyses suggest that cancers with high cytoplasmic ezrin have a more invasive phenotype. This study supports our previous findings associating cytoplasmic ezrin with more aggressive behavior and poorer outcome and indicates the need for a multi-institutional study to validate the use of cytoplasmic ezrin as a biomarker for treatment planning in head and neck squamous cell carcinoma.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cytoskeletal Proteins/analysis , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Microfilament Proteins/analysis , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Cytoplasm/metabolism , Female , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Tissue Array Analysis
14.
J Pathol ; 226(1): 108-19, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21953071

ABSTRACT

Invasive lobular carcinoma (ILC) of the breast, characterized by loss of E-cadherin expression, accounts for 5-15% of invasive breast cancers and it is believed to arise via a linear histological progression. Genomic studies have identified a clonal relationship between ILC and concurrent lobular carcinoma in situ (LCIS) lesions, suggesting that LCIS may be a precursor lesion. It has been shown that an LCIS diagnosis confers a 15-20% risk of progression to ILC over a lifetime. Currently no molecular test or markers can identify LCIS lesions likely to progress to ILC. Since microRNA (miRNA) expression changes have been detected in a number of other cancer types, we explored whether their dysregulation might be detected during progression from LCIS to ILC. Using the Illumina miRNA profiling platform, designed for simultaneous analysis of 470 mature miRNAs, we analysed the profiles of archived normal breast epithelium, LCIS lesions found alone, LCIS lesions concurrent with ILC, and the concurrent ILCs as a model of linear histological progression towards ILC. We identified two sets of differentially expressed miRNAs, the first set highly expressed in normal epithelium, including hsa-miR-224, -139, -10b, -450, 140, and -365, and the second set up-regulated during lobular neoplasia progression, including hsa-miR-375, -203, -425-5p, -183, -565, and -182. Using quantitative RT-PCR, we validated a trend of increasing expression for hsa-miR-375, hsa-miR-182, and hsa-miR-183 correlating with ILC progression. As we detected increased expression of hsa-miR-375 in LCIS lesions synchronous with ILC, we sought to determine whether hsa-miR-375 might induce phenotypes reminiscent of lobular neoplasia by expressing it in the MCF-10A 3D culture model of mammary acinar morphogenesis. Increased expression of hsa-miR-375 resulted in loss of cellular organization and acquisition of a hyperplastic phenotype. These data suggest that dysregulated miRNA expression contributes to lobular neoplastic progression.


Subject(s)
Acinar Cells/pathology , Breast Neoplasms/genetics , Carcinoma in Situ/genetics , Carcinoma, Lobular/genetics , Cell Polarity/genetics , MicroRNAs/genetics , Breast Neoplasms/pathology , Carcinoma in Situ/pathology , Carcinoma, Lobular/pathology , Disease Progression , Female , Fluorescent Antibody Technique , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Microdissection , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA
15.
J Enzyme Inhib Med Chem ; 25(3): 383-90, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19857049

ABSTRACT

The mechanochemical synthesis and characterization of a zinc complex with famotidine is described. The complex was characterized by microanalysis and a number of spectroscopic techniques. The complex was of M:L dihydrate type. Derivatization of famotidine with zinc appears to enhance the activity of the drug by inhibiting the growth of Helicobacter pylori (two reference and 34 clinical isolates). The complex inhibited the growth of H. pylori in an MIC range of 1-8 microg mL(-1). The anti-H. pylori activity of the zinc-famotidine complex against antibiotic-resistant strains was nearly comparable to that of antibiotic-susceptible strains. The complex was found to be far less toxic than the parent drug, as demonstrated by its higher LD(50) value. In the human urease enzyme inhibition assay the complex exhibited significant inhibition. The new complex appears to be more useful in eradicating both the antibiotic-susceptible and antibiotic-resistant strains of H. pylori.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Famotidine/chemistry , Helicobacter pylori/drug effects , Urease/antagonists & inhibitors , Zinc/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Ulcer Agents , Drug Resistance, Microbial , Famotidine/pharmacology , Humans , Microbial Sensitivity Tests
16.
Proc Natl Acad Sci U S A ; 105(39): 14867-72, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18806226

ABSTRACT

The active acquisition of epigenetic changes is a poorly understood but important process in development, differentiation, and disease. Our work has shown that repression of the p16/pRb pathway in human epithelial cells, a condition common to stem cells and many tumor cells, induces dynamic epigenetic remodeling resulting in the targeted methylation of a selected group of CpG islands. We hypothesized that cells in this epigenetically plastic state could be programmed by the microenvironment to acquire epigenetic changes associated with tumorigenesis. Here, we describe an in vitro model system where epigenetically plastic cells were placed in an environment that induced epithelial to mesenchymal transition (EMT) and led to a program of acquired de novo DNA methylation at targeted sites. In this model, we found that repression of E-cadherin transcription preceded the subsequent acquisition of methylated CpG sites. Furthermore, the induction of EMT was accompanied by de novo methylation of several other gene promoters, including those of the estrogen receptor and Twist. These data demonstrate that signals from the microenvironment can induce phenotypic and gene expression changes associated with targeted de novo epigenetic alterations important in tumor progression, and that these alterations occur through a deterministic, rather than stochastic, mechanism. Given the dynamic epigenetic reprogramming that occurs in these cells, DNA methylation profiles observed in human tumors may reflect the history of environmental exposures during the genesis of a tumor.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , DNA Methylation , Epithelial Cells/pathology , Humans , Mesoderm/pathology , Promoter Regions, Genetic , Serum , Smad2 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
17.
Clin Cancer Res ; 13(22 Pt 1): 6834-41, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006786

ABSTRACT

PURPOSE: p16(INK4a) has been appreciated as a key regulator of cell cycle progression and senescence. Cultured human mammary epithelial cells that lack p16(INK4a) activity have been shown to exhibit premalignant phenotypes, such as telomeric dysfunction, centrosomal dysfunction, a sustained stress response, and, most recently, a dysregulation of chromatin remodeling and DNA methylation. These data suggest that cells that lack p16(INK4a) activity would be at high risk for breast cancer development and may exhibit an increased frequency of DNA methylation events in early cancer. EXPERIMENTAL DESIGN: To test this hypothesis, the frequencies of INK4a/ARF promoter hypermethylation, as well as four additional selected loci, were tested in the initial random periareolar fine needle aspiration samples from 86 asymptomatic women at high risk for development of breast cancer, stratified using the Masood cytology index. RESULTS: INK4a/ARF promoter hypermethylation was observed throughout all early stages of intraepithelial neoplasia and, importantly, in morphologically normal-appearing mammary epithelial cells; 29 of 86 subjects showed INK4a/ARF promoter hypermethylation in at least one breast. Importantly, INK4a/ARF promoter hypermethylation was not associated with atypia, and the frequency of hypermethylation did not increase with increasing Masood cytology score. The frequency of INK4a/ARF promoter hypermethylation was associated with the combined frequency of promoter hypermethylation of retinoic acid receptor-beta2, estrogen receptor-alpha, and breast cancer-associated 1 genes (P = 0.001). CONCLUSIONS: Because INK4a/ARF promoter hypermethylation does not increase with age but increases with the frequency of other methylation events, we predict that INK4a/ARF promoter hypermethylation may serve as a marker of global methylation dysregulation.


Subject(s)
Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Gene Silencing , Mammary Glands, Human/metabolism , Adult , Aged , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Mammary Glands, Human/cytology , Middle Aged , Promoter Regions, Genetic , Risk
18.
J Biol Chem ; 281(34): 24790-802, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16766534

ABSTRACT

Alterations in DNA methylation are important in cancer, but the acquisition of these alterations is poorly understood. Using an unbiased global screen for CpG island methylation events, we have identified a non-random pattern of DNA hypermethylation acquired in p16-repressed cells. Interestingly, this pattern included loci located upstream of a number of homeobox genes. Upon removal of p16(INK4A) activity in primary human mammary epithelial cells, polycomb repressors, EZH2 and SUZ12, are up-regulated and recruited to HOXA9, a locus expressed during normal breast development and epigenetically silenced in breast cancer. We demonstrate that at this targeted locus, the up-regulation of polycomb repressors is accompanied by the recruitment of DNA methyltransferases and the hypermethylation of DNA, an endpoint, which we show to be dependent on SUZ12 expression. These results demonstrate a causal role of p16(INK4A) disruption in modulating DNA hypermethylation, and identify a dynamic and active process whereby epigenetic modulation of gene expression is activated as an early event in breast tumor progression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Silencing , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Neoplasm Proteins , Nuclear Proteins , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Colloids Surf B Biointerfaces ; 46(4): 204-17, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16337779

ABSTRACT

Phosphorylcholine (PC) moieties were chemically attached to surfaces of polymer microparticles by addition of 2-methylacryloyloxyethyl phosphorylcholine monomer to the seeded, semi-continuous polymerisations of methyl methacrylate (MMA) and butyl acrylate (BA). The surface of the bio-functionalised polymer microparticles was principally characterised using X-ray photoelectron spectroscopy (XPS), dynamic nuclear magnetic resonance (NMR) spectroscopy, scanning electron microscopy (SEM), photon correlation spectroscopy (PCS), acoustophoresis and enzyme-linked immunosorbent assays (ELISA). It was found that the persulphate initiating species are concealed behind the phosphorylcholine containing monomer sequence located on the surface of the microparticles. The combination of analytical techniques showed that the surfaces of the polymer microparticles are extremely mobile above the glass transition temperature of the co-polymer and able to rearrange depending on the environment in which they are placed. This allows the phosphorylcholine moiety to be preferentially expressed at the surface in aqueous media, but not so in the dry state or conditions of ultra-high vacuum. In terms of the nature of the biocompatibility of phosphorylcholine containing polymers, no evidence was found for the irreversible structuring of water molecules around the phosphorylcholine moiety in the wet state. The results of this work suggest that a more likely contributory reason for the protein-resistant nature of phosphorylcholine containing polymers is the mobility of the phosphorylcholine moiety. Increases in biocompatibility correspond with increases in the hydrophilicity of a polymer surface when phosphorylcholine is preferentially expressed. A large free water fraction may be present in the phosphorylcholine containing monomer sequence, as part of a hydrogel structure located at the surface of the polymer microparticles. This, coupled with concomitant modification of the local electrical double-layer very close to the surface may also play a critical role in reducing protein-surface interactions.


Subject(s)
Biocompatible Materials/chemistry , Phosphorylcholine/chemistry , Polymers/chemistry , Chemical Phenomena , Chemistry, Physical , Enzyme-Linked Immunosorbent Assay , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Materials Testing/methods , Microscopy, Electron, Scanning , Molecular Structure , Particle Size , Spectrometry, Fluorescence , Spectrometry, X-Ray Emission , Surface Properties
20.
Genes Dev ; 17(17): 2094-107, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12923058

ABSTRACT

Desmoplastic small round cell tumor (DSRCT) is defined by a chimeric transcription factor, resulting from fusion of the N-terminal domain of the Ewing's sarcoma gene EWS to the three C-terminal zinc fingers of the Wilms' tumor suppressor WT1. Although DNA-binding sites have been defined for the uninterrupted WT1 zinc finger domains, the most prevalent isoforms of both WT1 and EWS-WT1 have an insertion of three amino acids [lysine, threonine, and serine (KTS)], which abrogates binding to known consensus sequences and transactivation of known target genes. Here, we used cDNA subtractive hybridization to identify an endogenous gene, LRRC15, which is specifically up-regulated after inducible expression of EWS-WT1(+KTS) in cancer cell lines, and is expressed within primary DSRCT cells. The chimeric protein binds in vitro and in vivo to a specific element upstream of LRRC15, leading to dramatic transcriptional activation. Mutagenesis studies define the optimal binding site of the (+KTS) isoform of EWS-WT1 as 5'-GGAGG(A/G)-3'. LRRC15 encodes a leucine-rich transmembrane protein, present at the leading edge of migrating cells, the expression of which in normal tissues is restricted to the invasive cytotrophoblast layer of the placenta; small interfering (siRNA)-mediated suppression of LRRC15 expression in breast cancer cells leads to abrogation of invasiveness in vitro. Together, these observations define the consequence of (KTS) insertion within WT1-derived zinc fingers, and identify a novel EWS-WT1 transcriptional target implicated in tumor invasiveness.


Subject(s)
DNA/metabolism , Oncogene Proteins, Fusion/metabolism , RNA Processing, Post-Transcriptional/physiology , Amino Acid Sequence , Binding Sites/physiology , Humans , Molecular Sequence Data , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...