Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(17): 24284-302, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27015368

ABSTRACT

Loss of tumor suppressor proteins, such as the retinoblastoma protein (Rb), results in tumor progression and metastasis. Metastasis is facilitated by low oxygen availability within the tumor that is detected by hypoxia inducible factors (HIFs). The HIF1 complex, HIF1α and dimerization partner the aryl hydrocarbon receptor nuclear translocator (ARNT), is the master regulator of the hypoxic response. Previously, we demonstrated that Rb represses the transcriptional response to hypoxia by virtue of its association with HIF1. In this report, we further characterized the role Rb plays in mediating hypoxia-regulated genetic programs by stably ablating Rb expression with retrovirally-introduced short hairpin RNA in LNCaP and 22Rv1 human prostate cancer cells. DNA microarray analysis revealed that loss of Rb in conjunction with hypoxia leads to aberrant expression of hypoxia-regulated genetic programs that increase cell invasion and promote neuroendocrine differentiation. For the first time, we have established a direct link between hypoxic tumor environments, Rb inactivation and progression to late stage metastatic neuroendocrine prostate cancer. Understanding the molecular pathways responsible for progression of benign prostate tumors to metastasized and lethal forms will aid in the development of more effective prostate cancer therapies.


Subject(s)
Biomarkers, Tumor/genetics , Cell Differentiation , Hypoxia/genetics , Neuroendocrine Cells/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Retinoblastoma Protein/metabolism , Apoptosis , Cell Movement , Cell Proliferation , Gene Expression Profiling , Gene Regulatory Networks , Humans , Male , Neoplasm Invasiveness , Neuroendocrine Cells/metabolism , Prostatic Neoplasms/metabolism , Retinoblastoma Protein/genetics , Tumor Cells, Cultured
2.
PLoS One ; 9(6): e99214, 2014.
Article in English | MEDLINE | ID: mdl-24919196

ABSTRACT

Localized hypoxia in solid tumors activates transcriptional programs that promote the metastatic transformation of cells. Like hypoxia-inducible hyper-vascularization, loss of the retinoblastoma protein (Rb) is a trait common to advanced stages of tumor progression in many metastatic cancers. However, no link between the role of Rb and hypoxia-driven metastatic processes has been established. We demonstrated that Rb is a key mediator of the hypoxic response mediated by HIF1α/ß, the master regulator of the hypoxia response, and its essential co-activator, the thyroid hormone receptor/retinoblastoma-interacting protein (TRIP230). Furthermore, loss of Rb unmasks the full co-activation potential of TRIP230. Using small inhibitory RNA approaches in vivo, we established that Rb attenuates the normal physiological response to hypoxia by HIF1α. Notably, loss of Rb results in hypoxia-dependent biochemical changes that promote acquisition of an invasive phenotype in MCF7 breast cancer cells. In addition, Rb is present in HIF1α-ARNT/HIF1ß transcriptional complexes associated with TRIP230 as determined by co-immuno-precipitation, GST-pull-down and ChIP assays. These results demonstrate that Rb is a negative modulator of hypoxia-regulated transcription by virtue of its direct effects on the HIF1 complex. This work represents the first link between the functional ablation of Rb in tumor cells and HIF1α-dependent transcriptional activation and invasion.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Cytoskeletal Proteins , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , MCF-7 Cells , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , Retinoblastoma Protein/genetics
3.
Proteomics Clin Appl ; 6(9-10): 476-85, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22930592

ABSTRACT

PURPOSE: This proteomics study was designed to determine the utility of iTRAQ MALDI-TOF/TOF technology to compare plasma samples from carefully phenotyped mild, atopic asthma subjects undergoing allergen inhalation challenge. EXPERIMENTAL DESIGN: Eight adult subjects with mild, allergic asthma (four early responders (ERs) and four dual responders (DRs)) participated in the allergen inhalation challenge. Blood samples were collected prior to and 2 h after the inhalation challenge. Sixteen plasma samples (two per subject), technical replicates, and pooled controls were analyzed using iTRAQ. Technical validation was performed using LC-MRM/MS. Moderated robust regression was used to determine differentially expressed proteins. RESULTS: Although this study did not show significant differences between pre- and post-challenge samples, discriminant analysis indicated that certain proteins responded differentially to allergen challenge with respect to responder type. At pre-challenge, fibronectin was significantly elevated in DRs compared to ERs and remained significant in the multiple reaction monitoring validation. CONCLUSIONS AND CLINICAL RELEVANCE: This proof of principle demonstration has shown that iTRAQ can uncover differences in the human plasma proteome between two endotypes of asthma and merits further application of iTRAQ to larger cohorts of asthma and other respiratory diseases.


Subject(s)
Asthma/blood , Proteomics , Administration, Inhalation , Adult , Allergens/immunology , Asthma/immunology , Asthma/pathology , Chromatography, High Pressure Liquid , Discriminant Analysis , Female , Fibronectins/blood , Humans , Male , Middle Aged , Proteome/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Young Adult
4.
PLoS One ; 7(1): e29545, 2012.
Article in English | MEDLINE | ID: mdl-22235307

ABSTRACT

The activated AHR/ARNT complex (AHRC) regulates the expression of target genes upon exposure to environmental contaminants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Importantly, evidence has shown that TCDD represses estrogen receptor (ER) target gene activation through the AHRC. Our data indicates that AHR and ARNT act independently from each other at non-dioxin response element sites. Therefore, we sought to determine the specific functions of AHR and ARNT in estrogen-dependent signaling in human MCF7 breast cancer and human ECC-1 endometrial carcinoma cells. Knockdown of AHR with siRNA abrogates dioxin-inducible repression of estrogen-dependent gene transcription. Intriguingly, knockdown of ARNT does not effect TCDD-mediated repression of estrogen-regulated transcription, suggesting that AHR represses ER function independently of ARNT. This theory is supported by the ability of the selective AHR modulator 3',4'-dimethoxy-α-naphthoflavone (DiMNF) to repress estrogen-inducible transcription. Furthermore, basal and estrogen-activated transcription of the genes encoding cathepsin-D and pS2 are down-regulated in MCF7 cells but up-regulated in ECC-1 cells in response to loss of ARNT. These responses are mirrored at the protein level with cathepsin-D. Furthermore, knock-down of ARNT led to opposite but corresponding changes in estrogen-stimulated proliferation in both MCF7 and ECC-1 cells. We have obtained experimental evidence demonstrating a dioxin-dependent repressor function for AHR and a dioxin-independent co-activator/co-repressor function for ARNT in estrogen signalling. These results provide us with further insight into the mechanisms of transcription factor crosstalk and putative therapeutic targets in estrogen-positive cancers.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Estrogens/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Aryl Hydrocarbon Receptor Nuclear Translocator/deficiency , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Gene Knockdown Techniques , Humans , Phenotype , Polychlorinated Dibenzodioxins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects
5.
J Pharmacol Exp Ther ; 335(2): 273-83, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20716622

ABSTRACT

The putative cardioprotective and chemopreventive properties of the red wine phenolic resveratrol (RES) have made it the subject of a growing body of clinical and basic research. We have begun investigations focusing on the effects of RES on the activity of the aryl hydrocarbon receptor (AHR) complex. Our evidence suggests that RES is a potent repressor of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible gene transcription in estrogen receptor (ER)-positive human breast, lung, and colon cancer cell lines. RES activates the transcription of the ER target genes to the same degree as estradiol (E(2)) in human MCF-7 breast cancer cells. Unlike E(2), which can only diminish TCDD-inducible CYP1A1 gene transcription by approximately 50%, RES can completely abrogate this response. Furthermore, 50% repression of TCDD-inducible transcription can be achieved with 100 nM RES, approximately 2.5 orders of magnitude lower than concentrations required for maximal inhibition, suggesting that multiple mechanisms are responsible for this effect. RES (100 nM) does not prevent ligand binding of a TCDD analog, nor does it prevent AHR from binding to its response element in the 5'-regulatory region of the CYP1A1 gene. Small inhibitory RNAs directed to ERα have demonstrated that RES-mediated repression of CYP1A1 depends on ERα. Whereas CYP1A1 protein levels in MCF-7 cells are refractory to the low-dose transcriptional effects of RES, a concomitant decrease in CYP1A1 protein levels is observed in Caco-2 cells. These results highlight a low-dose RES effect that could occur at nutritionally relevant exposures and are distinct from the high-dose effects often characterized.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Estrogen Receptor alpha/biosynthesis , Protective Agents/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Stilbenes/pharmacology , Binding, Competitive , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Cytochrome P-450 CYP1A1/genetics , Dose-Response Relationship, Drug , Estrogen Receptor alpha/genetics , Gene Expression/drug effects , Humans , Ligands , Promoter Regions, Genetic/drug effects , Protective Agents/administration & dosage , Protein Binding , RNA Interference , Radioligand Assay , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Stilbenes/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...