Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Breast Cancer Res ; 22(1): 122, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33148314

ABSTRACT

BACKGROUND: The role of nuclear receptors in both the aetiology and treatment of breast cancer is exemplified by the use of the oestrogen receptor (ER) as a prognostic marker and treatment target. Treatments targeting the oestrogen signalling pathway are initially highly effective for most patients. However, for the breast cancers that fail to respond, or become resistant, to current endocrine treatments, the long-term outlook is poor. ER is a member of the nuclear receptor superfamily, comprising 48 members in the human, many of which are expressed in the breast and could be used as alternative targets in cases where current treatments are ineffective. METHODS: We used sparse canonical correlation analysis to interrogate potential novel nuclear receptor expression relationships in normal breast and breast cancer. These were further explored using whole transcriptome profiling in breast cancer cells after combinations of ligand treatments. RESULTS: Using this approach, we discovered a tumour suppressive relationship between the mineralocorticoid receptor (MR) and retinoic acid receptors (RAR), in particular RARß. Expression profiling of MR expressing breast cancer cells revealed that mineralocorticoid and retinoid co-treatment activated an expression program consistent with a reverse Warburg effect and growth inhibition, which was not observed with either ligand alone. Moreover, high expression of both MR and RARB was associated with improved breast cancer-specific survival. CONCLUSION: Our study reveals a previously unknown relationship between MR and RAR in the breast, which is dependent on menopausal state and altered in malignancy. This finding identifies potential new targets for the treatment of breast cancers that are refractory to existing therapeutic options.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Receptors, Mineralocorticoid/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction , Warburg Effect, Oncologic , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Computational Biology , Female , Humans , Receptors, Estrogen/metabolism , Receptors, Mineralocorticoid/genetics , Survival Rate , Tumor Cells, Cultured
2.
Dev Cell ; 49(2): 279-292.e5, 2019 04 22.
Article in English | MEDLINE | ID: mdl-31014480

ABSTRACT

The correct assignment of cell fate within fields of multipotent progenitors is essential for accurate tissue diversification. The first lymphatic vessels arise from pre-existing veins after venous endothelial cells become specified as lymphatic progenitors. Prox1 specifies lymphatic fate and labels these progenitors; however, the mechanisms restricting Prox1 expression and limiting the progenitor pool remain unknown. We identified a zebrafish mutant that displayed premature, expanded, and prolonged lymphatic specification. The gene responsible encodes the regulator of alternative splicing, Nova2. In zebrafish and human endothelial cells, Nova2 selectively regulates pre-mRNA splicing for components of signaling pathways and phosphoproteins. Nova2-deficient endothelial cells display increased Mapk/Erk signaling, and Prox1 expression is dynamically controlled by Erk signaling. We identify a mechanism whereby Nova2-regulated splicing constrains Erk signaling, thus limiting lymphatic progenitor cell specification. This identifies the capacity of a factor that tunes mRNA splicing to control assignment of cell fate during vascular differentiation.


Subject(s)
Lymphatic Vessels/metabolism , MAP Kinase Signaling System , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Alternative Splicing , Animals , Cell Differentiation , Cell Lineage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Homeodomain Proteins/metabolism , Humans , Lymphangiogenesis , Lymphatic Vessels/cytology , Male , Neuro-Oncological Ventral Antigen , Tumor Suppressor Proteins/metabolism , Veins/cytology , Veins/metabolism , Zebrafish
3.
J Mol Endocrinol ; 62(3): R223-R237, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30620710

ABSTRACT

Breast cancer is a heterogeneous disease, and the complexity of breast carcinogenesis is associated with epigenetic modification. There are several major classes of epigenetic enzymes that regulate chromatin activity. This review will focus on the nine mammalian protein arginine methyltransferases (PRMTs) and the dysregulation of PRMT expression and function in breast cancer. This class of enzymes catalyse the mono- and (symmetric and asymmetric) di-methylation of arginine residues on histone and non-histone target proteins. PRMT signalling (and R methylation) drives cellular proliferation, cell invasion and metastasis, targeting (i) nuclear hormone receptor signalling, (ii) tumour suppressors, (iii) TGF-ß and EMT signalling and (iv) alternative splicing and DNA/chromatin stability, influencing the clinical and survival outcomes in breast cancer. Emerging reports suggest that PRMTs are also implicated in the development of drug/endocrine resistance providing another prospective avenue for the treatment of hormone resistance and associated metastasis. The complexity of PRMT signalling is further underscored by the degree of alternative splicing and the scope of variant isoforms (with distinct properties) within each PRMT family member. The evolution of PRMT inhibitors, and the ongoing clinical trials of PRMT inhibitors against a subgroup of solid cancers, coupled to the track record of lysine methyltransferases inhibitors in phase I/II clinical trials against cancer underscores the potential therapeutic utility of targeting PRMT epigenetic enzymes to improve survival outcomes in aggressive and metastatic breast cancer.


Subject(s)
Arginine/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic/genetics , Animals , Female , Humans , Methylation
4.
Exerc Sport Sci Rev ; 46(2): 97-104, 2018 04.
Article in English | MEDLINE | ID: mdl-29346164

ABSTRACT

Exercise induces various physical and metabolic changes in skeletal muscle that adaptively reprograms this tissue to current physiological and environmental demands. Underlying these changes are broad modifications to gene expression. We postulate that the nuclear hormone receptor, Nor-1, is activated after exercise, and this transcription factor modifies gene expression to drive the molecular and cellular adaptations associated with contractile reorganization.


Subject(s)
Adaptation, Physiological , Exercise/physiology , Muscle, Skeletal/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 3/metabolism , Animals , Autophagy , Calcium Signaling , Gene Expression , Humans , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/blood supply , Muscle, Skeletal/physiology , Sarcomeres/metabolism
5.
EBioMedicine ; 16: 63-75, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28119061

ABSTRACT

While invasion and metastasis of tumour cells are the principle factor responsible for cancer related deaths, the mechanisms governing the process remain poorly defined. Moreover, phenotypic divergence of sub-populations of tumour cells is known to underpin alternative behaviors linked to tumour progression such as proliferation, survival and invasion. In the context of melanoma, heterogeneity between two transcription factors, BRN2 and MITF, has been associated with phenotypic switching between predominantly invasive and proliferative behaviors respectively. Epigenetic changes, in response to external cues, have been proposed to underpin this process, however the mechanism by which the phenotypic switch occurs is unclear. Here we report the identification of the NFIB transcription factor as a novel downstream effector of BRN2 function in melanoma cells linked to the migratory and invasive characteristics of these cells. Furthermore, the function of NFIB appears to drive an invasive phenotype through an epigenetic mechanism achieved via the upregulation of the polycomb group protein EZH2. A notable target of NFIB mediated up-regulation of EZH2 is decreased MITF expression, which further promotes a less proliferative, more invasive phenotype. Together our data reveal that NFIB has the ability to promote dynamic changes in the chromatin state of melanoma cells to facilitate migration, invasion and metastasis.


Subject(s)
Cell Movement/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Homeodomain Proteins/genetics , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , NFI Transcription Factors/genetics , POU Domain Factors/genetics , Animals , Blotting, Western , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Mice, Inbred BALB C , Mice, Knockout , Microphthalmia-Associated Transcription Factor/metabolism , Microscopy, Fluorescence , NFI Transcription Factors/metabolism , Neoplasm Invasiveness , POU Domain Factors/metabolism , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
6.
Endocrinology ; 158(3): 431-447, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27983866

ABSTRACT

Breast cancer is a heterogeneous disease and its complexity has hindered the development of efficacious treatments targeting all breast cancer subtypes. Many studies have linked the diversity of breast carcinogenesis and metastasis to aberrant epigenetic signaling and control. Here, we focus on the current state of the discipline and review the major epigenetic enzymes controlling chromatin structure and function in the context of breast cancer, including (1) DNA methyltransferases, (2) lysine methyltransferases and demethylases, (3) protein arginine methyltransferases, and (4) histone acetyltransferases and deacetylases. Moreover, therapeutic drugs targeting these epigenetic enzymes are rapidly emerging and/or undergoing clinical trials. Therefore, we discuss the pharmacological manipulation of epigenetic enzymes for breast cancer treatment and present new clinical and survival outcome analysis on epigenetic factors that have evaded analysis to date. Understanding and pharmacologically exploiting epigenetic regulation in breast cancer promises to be an essential aspect of next-generation drug development and adjuvant therapies targeting advanced disease and treatment-resistant tumors.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Epigenesis, Genetic , Animals , Carcinogenesis , DNA Modification Methylases/metabolism , Female , Histone Deacetylases/metabolism , Histone Demethylases/metabolism , Humans , Lysine Acetyltransferases/metabolism
7.
EBioMedicine ; 11: 101-117, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27568222

ABSTRACT

RORα is a member of the nuclear receptor (NR) superfamily and analysis of the (global) RORα-deficient mouse model revealed this NR has a role in glycemic control and fat deposition. Therefore, we generated an adipose-specific RORα 'gain of function' mouse model under the control of the fatty acid binding protein 4 (FABP4) promoter to elucidate the function of RORα in adipose tissue. The Tg-FABP4-RORα4 mice demonstrated a shift in fat distribution to non-adipose tissues when challenged with a high fat diet (HFD). Specifically, we observed a subcutaneous lipodystrophy, accompanied by hepatomegaly (fatty liver/mild portal fibrosis) and splenomegaly; in a background of decreased weight gain and total body fat after HFD. Moreover, we observed significantly higher fasting blood glucose and impaired clearance of glucose in Tg-FABP4-RORα4 mice. Genome wide expression and qPCR profiling analysis identified: (i) subcutaneous adipose specific decreases in the expression of genes involved in fatty acid biosynthesis, lipid droplet expansion and glycemic control, and (ii) the fibrosis pathway as the most significant pathway [including dysregulation of the collagen/extracellular matrix (ECM) pathways] in subcutaneous adipose and liver. The pathology presented in the Tg-FABP4-RORα4 mice is reminiscent of human metabolic disease (associated with aberrant ECM expression) highlighting the therapeutic potential of this NR.


Subject(s)
Adipose Tissue/metabolism , Adiposity/genetics , Blood Glucose , Gene Expression , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Adipose Tissue/immunology , Adipose Tissue/pathology , Adiposity/immunology , Animals , Biomarkers , Cluster Analysis , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Profiling , Genotype , Glucose Tolerance Test , Hepatomegaly/genetics , Hepatomegaly/metabolism , Hepatomegaly/pathology , Humans , Insulin Resistance , Lipid Metabolism , Lipids/blood , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Organ Specificity , Phenotype , Splenomegaly/genetics , Splenomegaly/metabolism , Splenomegaly/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transgenes , Weight Gain
8.
Mol Endocrinol ; 30(6): 660-76, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27144290

ABSTRACT

Skeletal muscle remodels metabolic capacity, contractile and exercise phenotype in response to physiological demands. This adaptive remodeling response to physical activity can ameliorate/prevent diseases associated with poor diet and lifestyle. Our previous work demonstrated that skeletal muscle-specific transgenic expression of the neuron-derived orphan nuclear receptor, Nor-1 drives muscle reprogramming, improves exercise endurance, and oxidative metabolism. The current manuscript investigates the association between exercise, Nor-1 expression and the role of Nor-1 in adaptive remodeling. We demonstrate that Nor-1 expression is induced by exercise and is dependent on calcium/calcineurin signaling (in vitro and in vivo). Analysis of fatigue-resistant transgenic mice that express Nor-1 in skeletal muscle revealed increased hypertrophy and vascularization of muscle tissue. Moreover, we demonstrate that transgenic Nor-1 expression is associated with increased intracellular recycling, ie, autophagy, involving 1) increased expression of light chain 3A or LC3A-II, autophagy protein 5, and autophagy protein 12 in quadriceps femoris muscle extracts from Tg-Nor-1 (relative to Wild-type (WT) littermates); 2) decreased p62 expression indicative of increased autophagolysosome assembly; and 3) decreased mammalian target of rapamycin complex 1 activity. Transfection of LC3A-GFP-RFP chimeric plasmid demonstrated that autophagolysosome formation was significantly increased by Nor-1 expression. Furthermore, we demonstrated a single bout of exercise induced LC3A-II expression in skeletal muscle from C57BL/6 WT mice. This study, when combined with our previous studies, demonstrates that Nor-1 expression drives multiple physiological changes/pathways that are critical to the beneficial responses of muscle to exercise and provides insights into potential pharmacological manipulation of muscle reprogramming for the treatment of lifestyle induced chronic diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Physical Conditioning, Animal , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Calcineurin/metabolism , Calcium/metabolism , Cell Line , DNA-Binding Proteins/genetics , Hypertrophy , Lysosomes/drug effects , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Models, Biological , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/parasitology , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction/drug effects , Sirolimus/pharmacology
9.
EBioMedicine ; 6: 59-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27211549

ABSTRACT

We have previously reported that RORγ expression was decreased in ER-ve breast cancer, and increased expression improves clinical outcomes. However, the underlying RORγ dependent mechanisms that repress breast carcinogenesis have not been elucidated. Here we report that RORγ negatively regulates the oncogenic TGF-ß/EMT and mammary stem cell (MaSC) pathways, whereas RORγ positively regulates DNA-repair. We demonstrate that RORγ expression is: (i) decreased in basal-like subtype cancers, and (ii) inversely correlated with histological grade and drivers of carcinogenesis in breast cancer cohorts. Furthermore, integration of RNA-seq and ChIP-chip data reveals that RORγ regulates the expression of many genes involved in TGF-ß/EMT-signaling, DNA-repair and MaSC pathways (including the non-coding RNA, LINC00511). In accordance, pharmacological studies demonstrate that an RORγ agonist suppresses breast cancer cell viability, migration, the EMT transition (microsphere outgrowth) and mammosphere-growth. In contrast, RNA-seq demonstrates an RORγ inverse agonist induces TGF-ß/EMT-signaling. These findings suggest pharmacological modulation of RORγ activity may have utility in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Repair , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Transforming Growth Factor beta/genetics , Benzamides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Metastasis , Piperazines/pharmacology , Propanols/pharmacology , Sequence Analysis, RNA , Signal Transduction
10.
PLoS One ; 11(1): e0147179, 2016.
Article in English | MEDLINE | ID: mdl-26812621

ABSTRACT

Nuclear hormone receptors have important roles in the regulation of metabolic and inflammatory pathways. The retinoid-related orphan receptor alpha (Rorα)-deficient staggerer (sg/sg) mice display several phenotypes indicative of aberrant lipid metabolism, including dyslipidemia, and increased susceptibility to atherosclerosis. In this study we demonstrate that macrophages from sg/sg mice have increased ability to accumulate lipids and accordingly exhibit larger lipid droplets (LD). We have previously shown that BMMs from sg/sg mice have significantly decreased expression of cholesterol 25-hydroxylase (Ch25h) mRNA, the enzyme that produces the oxysterol, 25-hydroxycholesterol (25HC), and now confirm this at the protein level. 25HC functions as an inverse agonist for RORα. siRNA knockdown of Ch25h in macrophages up-regulates Vldlr mRNA expression and causes increased accumulation of LDs. Treatment with physiological concentrations of 25HC in sg/sg macrophages restored lipid accumulation back to normal levels. Thus, 25HC and RORα signify a new pathway involved in the regulation of lipid homeostasis in macrophages, potentially via increased uptake of lipid which is suggested by mRNA expression changes in Vldlr and other related genes.


Subject(s)
Hydroxycholesterols/metabolism , Lipid Droplets/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Chromatography, Thin Layer , Drug Inverse Agonism , Lipid Metabolism , Lipids/analysis , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/chemistry , RNA Interference , RNA, Messenger/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Steroid Hydroxylases/antagonists & inhibitors , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Up-Regulation
11.
Am J Physiol Endocrinol Metab ; 308(2): E159-71, 2015 01 15.
Article in English | MEDLINE | ID: mdl-25424999

ABSTRACT

The Rar-related orphan receptor-α (Rorα) is a nuclear receptor that regulates adiposity and is a potential regulator of energy homeostasis. We have demonstrated that the Rorα-deficient staggerer (sg/sg) mice display a lean and obesity-resistant phenotype. Adaptive Ucp1-dependent thermogenesis in beige/brite and brown adipose tissue serves as a mechanism to increase energy expenditure and resist obesity. DEXA and MRI analysis demonstrated significantly decreased total fat mass and fat/lean mass tissue ratio in male chow-fed sg/sg mice relative to wt mice. In addition, we observed increased Ucp1 expression in brown adipose and subcutaneous white adipose tissue but not in visceral adipose tissue from Rorα-deficient mice. Moreover, this was associated with significant increases in the expression of the mRNAs encoding the thermogenic genes (i.e., markers of brown and beige adipose) Pparα, Errα, Dio2, Acot11/Bfit, Cpt1ß, and Cidea in the subcutaneous adipose in the sg/sg relative to WT mice. These changes in thermogenic gene expression involved the significantly increased expression of the (cell-fate controlling) histone-lysine N-methyltransferase 1 (Ehmt1), which stabilizes the Prdm16 transcriptional complex. Moreover, primary brown adipocytes from sg/sg mice displayed a higher metabolic rate, and further analysis was consistent with increased uncoupling. Finally, core body temperature analysis and infrared thermography demonstrated that the sg/sg mice maintained greater thermal control and cold tolerance relative to the WT littermates. We suggest that enhanced Ucp1 and thermogenic gene expression/activity may be an important contributor to the lean, obesity-resistant phenotype in Rorα-deficient mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Gene Expression Regulation/physiology , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Obesity/metabolism , Thermogenesis/physiology , Absorptiometry, Photon , Animals , Body Composition/physiology , Body Temperature/physiology , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Immunohistochemistry , Magnetic Resonance Imaging , Male , Mice , Mice, Neurologic Mutants , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Thermogenesis/genetics , Transcription Factors/metabolism , Uncoupling Protein 1
12.
Oncotarget ; 5(18): 8651-64, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25261374

ABSTRACT

Cumulative exposure to estrogen (E) and progesterone (P) over the menstrual cycle significantly influences the risk of developing breast cancer. Despite the dogma that PR in the breast merely serves as a marker of an active estrogen receptor (ER), and as an inhibitor of the proliferative actions of E, it is now clear that in the breast P increases proliferation independently of E action. We show here that the progesterone receptor (PR) and ER are expressed in different epithelial populations, and target non-overlapping pathways in the normal human breast. In breast cancer, PR becomes highly correlated with ER, and this convergence is associated with signaling pathways predictive of disease metastasis. These data challenge the established paradigm that ER and PR function co-operatively in normal breast, and have significant implications not only for our understanding of normal breast biology, but also for diagnosis, prognosis and/or treatment options in breast cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/secondary , Case-Control Studies , Cell Lineage , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Kaplan-Meier Estimate , Mammary Glands, Human/pathology , Prognosis , RNA, Messenger/metabolism , Receptor Cross-Talk , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Time Factors
13.
Mol Endocrinol ; 28(7): 1166-85, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24911119

ABSTRACT

Protein arginine methyltransferases (PRMTs) methylate arginine residues on histones and target transcription factors that play critical roles in many cellular processes, including gene transcription, mRNA splicing, proliferation, and differentiation. Recent studies have linked PRMT-dependent epigenetic marks and modifications to carcinogenesis and metastasis in cancer. However, the role of PRMT2-dependent signaling in breast cancer remains obscure. We demonstrate PRMT2 mRNA expression was significantly decreased in breast cancer relative to normal breast. Gene expression profiling, Ingenuity and protein-protein interaction network analysis after PRMT2-short interfering RNA transfection into MCF-7 cells, revealed that PRMT2-dependent gene expression is involved in cell-cycle regulation and checkpoint control, chromosomal instability, DNA repair, and carcinogenesis. For example, PRMT2 depletion achieved the following: 1) increased p21 and decreased cyclinD1 expression in (several) breast cancer cell lines, 2) decreased cell migration, 3) induced an increase in nucleotide excision repair and homologous recombination DNA repair, and 4) increased the probability of distance metastasis free survival (DMFS). The expression of PRMT2 and retinoid-related orphan receptor-γ (RORγ) is inversely correlated in estrogen receptor-positive breast cancer and increased RORγ expression increases DMFS. Furthermore, we found decreased expression of the PRMT2-dependent signature is significantly associated with increased probability of DMFS. Finally, weighted gene coexpression network analysis demonstrated a significant correlation between PRMT2-dependent genes and cell-cycle checkpoint, kinetochore, and DNA repair circuits. Strikingly, these PRMT2-dependent circuits are correlated with pan-cancer metagene signatures associated with epithelial-mesenchymal transition and chromosomal instability. This study demonstrates the role and significant correlation between a histone methyltransferase (PRMT2)-dependent signature, RORγ, the cell-cycle regulation, DNA repair circuits, and breast cancer survival outcomes.


Subject(s)
Breast Neoplasms/mortality , Gene Expression Regulation, Neoplastic/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Protein-Arginine N-Methyltransferases/genetics , Breast Neoplasms/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Chromosomal Instability/genetics , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DNA Breaks, Double-Stranded , DNA Repair/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Kinetochores/metabolism , MCF-7 Cells , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Protein Interaction Maps , Protein-Arginine N-Methyltransferases/biosynthesis , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Receptors, Estrogen/metabolism , Signal Transduction
14.
Mol Oncol ; 8(5): 998-1013, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24785096

ABSTRACT

Although molecular signatures based on transcript expression in breast cancer samples have provided new insights into breast cancer classification and prognosis, there are acknowledged limitations in current signatures. To provide rational, pathway-based signatures of disrupted physiology in cancer tissues that may be relevant to prognosis, this study has directly quantitated changed gene expression, between normal breast and cancer tissue, as a basis for signature development. The nuclear receptor (NR) family of transcription factors, and their coregulators, are fundamental regulators of every aspect of metazoan life, and were rigorously quantified in normal breast tissues and ERα positive and ERα negative breast cancers. Coregulator expression was highly correlated with that of selected NR in normal breast, particularly from postmenopausal women. These associations were markedly decreased in breast cancer, and the expression of the majority of coregulators was down-regulated in cancer tissues compared with normal. While in cancer the loss of NR-coregulator associations observed in normal breast was common, a small number of NR (Rev-ERBß, GR, NOR1, LRH-1 and PGR) acquired new associations with coregulators in cancer tissues. Elevated expression of these NR in cancers was associated with poorer outcome in large clinical cohorts, as well as suggesting the activation of ERα -related, but ERα-independent, pathways in ERα negative cancers. In addition, the combined expression of small numbers of NR and coregulators in breast cancer was identified as a signature predicting outcome in ERα negative breast cancer patients, not linked to proliferation and with predictive power superior to existing signatures containing many more genes. These findings highlight the power of predictive signatures derived from the quantitative determination of altered gene expression between normal breast and breast cancers. Taken together, the findings of this study identify networks of NR-coregulator associations active in normal breast but disrupted in breast cancer, and moreover provide evidence that signatures based on NR networks disrupted in cancer can provide important prognostic information in breast cancer patients.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Receptors, Cytoplasmic and Nuclear/genetics , Breast/metabolism , Breast/pathology , Female , Humans , Prognosis , Transcriptome
15.
Thyroid ; 24(6): 1000-11, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24559275

ABSTRACT

BACKGROUND: Nuclear receptors (NRs) play a key role in endocrine signaling and metabolism and are important therapeutic targets in a number of hormone-dependent malignancies. Studies on the role of NRs in thyroid cancer are limited. OBJECTIVE: The objective of the study was to examine systematically the expression of the 48 human NRs in a series of benign and malignant thyroid tissues. Within the papillary carcinoma cohort, we sought to determine if NR expression differed significantly by BRAF mutation status. PATIENTS AND METHODS: RNA was isolated from multinodular goiter (MNG; n=6), papillary carcinoma (PTC, n=14), follicular carcinoma (FC; n=5), and Hürthle cell carcinoma (HCC; n=7). The 48 human NRs were profiled in this panel by quantitative real time polymerase chain reaction. Protein expression for selected NRs (Rev-erbα and LXR-ß) was examined by immunohistochemistry (IHC) on tissue microarrays comprising benign and malignant thyroid tissues. RESULTS: Across all groups of benign and malignant thyroid tissue, there was prominent expression of LXR-ß and ROR-γ. Key findings in PTC were marked overexpression of RXR-γ and Rev-erbα compared to MNG. Within the PTC cohort, when BRAF(V600E) tumors were compared with wild type BRAF, there was relative upregulation of RXR-γ and Rev-erbα and downregulation of AR, ERR-γ, and ROR-γ. In FC, EAR-2 was overexpressed, while PPAR-α and PPAR-δ were underexpressed compared to MNG. The NR expression profile of HCC was distinct, characterized by significant downregulation of a wide range of NRs. IHC for Rev-erbα and LXR-ß localized protein expression to the tumor cells. Moderate to strong Rev-erbα immunostaining was seen in 22 out of 23 PTC, and, overall, staining was stronger than in the benign group. CONCLUSIONS: These results represent the first systematic examination of NR expression in thyroid cancer. Our finding of tumor-specific patterns of NR expression, as well as significant differences in NR expression between BRAF(V600E) and wild type BRAF PTC, provides a basis for further mechanistic studies and highlights potential novel therapeutic targets for this malignancy.


Subject(s)
Carcinoma, Papillary/physiopathology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Thyroid Gland/metabolism , Thyroid Neoplasms/physiopathology , Adenocarcinoma, Follicular/genetics , Adenoma, Oxyphilic , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Papillary/pathology , Cell Differentiation , Female , Goiter, Nodular/genetics , Humans , Male , Middle Aged , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/pathology , Transcriptome
16.
PLoS One ; 8(11): e78075, 2013.
Article in English | MEDLINE | ID: mdl-24223135

ABSTRACT

Ultraviolet radiation (UVR) is one of the most common mutagens encountered by humans and induces the formation of cyclobutane pyrimidine dimers (CPDs) and pyrimidine-(6-4)-pyrimidone photoproduct (6-4PP) lesions in the genomic DNA. To prevent the accumulation of deleterious mutations these lesions must be efficiently repaired, primarily by nucleotide excision repair. We have previously demonstrated that the NR4A family of nuclear receptors are crucial mediators of the DNA repair function of the MC1R signalling pathway in melanocytes. Here we explore the role of the NR4A2 protein in the DNA repair process further. Using EYFP tagged-NR4A2 we have demonstrated a UVR induced recruitment to distinct nuclear foci where they co-localise with known DNA repair proteins. We reveal that the N-terminal domain of the receptor is required for this translocation and identify a role for p38 and PARP signalling in this process. Moreover disruption of the functional integrity of the Ligand Binding Domain of the receptor by deleting the terminal helix 12 effectively blocks co-localisation of the receptor with DNA repair factors. Restored co-localisation of the mutant receptor with DNA repair proteins in the presence of a Histone Deacetylase Inhibitor suggests that impaired chromatin accessibility underpins the mis-localisation observed. Finally NR4A2 over-expression facilitated a more efficient clearance of UVR induced CPD and 6-4PP lesions. Taken together these data uncover a novel role for the NR4A nuclear receptors as direct facilitators of nucleotide excision repair.


Subject(s)
Cell Nucleus/metabolism , DNA Repair , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Ultraviolet Rays , Binding Sites , Cell Line, Tumor , Cell Nucleus/radiation effects , DNA Repair Enzymes/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , MAP Kinase Signaling System , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport
17.
Breast Cancer Res Treat ; 142(1): 211-23, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24122391

ABSTRACT

The interaction between breast tumor epithelial and stromal cells is vital for initial and recurrent tumor growth. While breast cancer-associated stromal cells provide a favorable environment for proliferation and metastasis, the molecular mechanisms contributing to this process are not fully understood. Nuclear receptors (NRs) are intracellular transcription factors that directly regulate gene expression. Little is known about the status of NRs in cancer-associated stroma. Nuclear Receptor Low-Density Taqman Arrays were used to compare the gene expression profiles of all 48 NR family members in a collection of primary cultured cancer-associated fibroblasts (CAFs) obtained from estrogen receptor (ER)α positive breast cancers (n = 9) and normal breast adipose fibroblasts (NAFs) (n = 7). Thirty-three of 48 NRs were expressed in both the groups, while 11 NRs were not detected in either. Three NRs (dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1 (DAX-1); estrogen-related receptor beta (ERR-ß); and RAR-related orphan receptor beta (ROR-ß)) were only detected in NAFs, while one NR (liver receptor homolog-1 (LRH-1)) was unique to CAFs. Of the NRs co-expressed, four were significantly down-regulated in CAFs compared with NAFs (RAR-related orphan receptor-α (ROR-α); Thyroid hormone receptor-ß (TR-ß); vitamin D receptor (VDR); and peroxisome proliferator-activated receptor-γ (PPAR-γ)). Quantitative immunohistochemistry for LRH-1, TR-ß, and PPAR-γ proteins in stromal fibroblasts from an independent panel of breast cancers (ER-positive (n = 15), ER-negative (n = 15), normal (n = 14)) positively correlated with mRNA expression profiles. The differentially expressed NRs identified in tumor stroma are key mediators in aromatase regulation and subsequent estrogen production. Our findings reveal a distinct pattern of NR expression that therefore fits with a sustained and increased local estrogen microenvironment in ER-positive tumors. NRs in CAFs may provide a new avenue for the development of intratumoral-targeted therapies in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Stromal Cells/metabolism , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Receptor, ErbB-2/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Risk Factors
18.
Mol Endocrinol ; 27(11): 1897-917, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24065705

ABSTRACT

The mRNA encoding Nor-1/NR4A3 is rapidly and strikingly induced by ß2-adrenergic signaling in glycolytic and oxidative skeletal muscle. In skeletal muscle cells, Nor-1 expression is important for the regulation of oxidative metabolism. Transgenic skeletal muscle-specific expression of activated Nor-1 resulted in the acquisition of an endurance phenotype, an increase in type IIA/X oxidative muscle fibers, and increased numbers of mitochondria. In the current study, we used dual-energy x-ray absorptiometry and magnetic resonance imaging analysis to demonstrate decreased adiposity in transgenic (Tg) Nor-1 mice relative to that in wild-type littermates. Furthermore, the Tg-Nor-1 mice were resistant to diet-induced weight gain and maintained fasting glucose at normoglycemic levels. Expression profiling and RT-quantitative PCR analysis revealed significant increases in genes involved in glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation, fatty acid oxidation, and glycogen synthesis, in concordance with the lean phenotype. Moreover, expression profiling identified several Z-disc and sarcomeric binding proteins that modulate fiber type phenotype and endurance, eg, α-actinin-3. In addition, we demonstrated that the Tg-Nor-1 mouse line has significantly higher glycogen content in skeletal muscle relative to that in wild-type littermates. Finally, we identified a decreased NAD(+)/NADH ratio with a concordant increase in peroxisome proliferator-activated receptor γ coactivator-1α1 protein/mRNA expression. Increased NADH was associated with an induction of the genes involved in the malate-aspartate shuttle and a decrease in the glycerol 3-phosphate shuttle, which maximizes aerobic ATP production. In conclusion, skeletal muscle-specific Nor-1 expression regulates genes and pathways that regulate adiposity, muscle fiber type metabolic capacity, and endurance.


Subject(s)
Adiposity , DNA-Binding Proteins/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Adipose Tissue/physiology , Animals , Carbohydrate Metabolism , DNA-Binding Proteins/genetics , Diet, High-Fat/adverse effects , Glycogen/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NAD/metabolism , Nerve Tissue Proteins/genetics , Obesity/etiology , Obesity/metabolism , Organ Specificity , Oxygen Consumption , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Endurance , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , Triglycerides/metabolism
19.
Cell Rep ; 4(2): 238-47, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23850288

ABSTRACT

Caveolae and caveolin-1 (CAV1) have been linked to several cellular functions. However, a model explaining their roles in mammalian tissues in vivo is lacking. Unbiased expression profiling in several tissues and cell types identified lipid metabolism as the main target affected by CAV1 deficiency. CAV1-/- mice exhibited impaired hepatic peroxisome proliferator-activated receptor α (PPARα)-dependent oxidative fatty acid metabolism and ketogenesis. Similar results were recapitulated in CAV1-deficient AML12 hepatocytes, suggesting at least a partial cell-autonomous role of hepatocyte CAV1 in metabolic adaptation to fasting. Finally, our experiments suggest that the hepatic phenotypes observed in CAV1-/- mice involve impaired PPARα ligand signaling and attenuated bile acid and FXRα signaling. These results demonstrate the significance of CAV1 in (1) hepatic lipid homeostasis and (2) nuclear hormone receptor (PPARα, FXRα, and SHP) and bile acid signaling.


Subject(s)
Bile Acids and Salts/metabolism , Caveolin 1/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Animals , Mice , Oxidation-Reduction , Signal Transduction
20.
IUBMB Life ; 65(8): 657-64, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23846999

ABSTRACT

Glycogen is an energy storage depot for the mammalian species. This review focuses on recent developments that have identified the role of nuclear hormone receptor (NR) signaling and epigenomic control in the regulation of important genes that modulate glycogen metabolism. Specifically, new studies have revealed that the NR4A subgroup (of the NR superfamily) are strikingly sensitive to beta-adrenergic stimulation in skeletal muscle, and transgenic studies in mice have revealed the expression of these NRs affects endurance and glycogen levels in muscle. Furthermore, other studies have demonstrated that one of the NR coregulator class of enzymes that mediate chromatin remodeling, the histone methyltransferases (for example, protein arginine methyltransferase 4) regulates the expression of several genes involved in glycogen metabolism and glycogen storage diseases in skeletal muscle. Importantly, NRs and histone methyltransferases, have the potential to be pharmacologically exploited and may provide novel targets in the quest to treat disorders of glycogen storage.


Subject(s)
Epigenomics , Glycogen/metabolism , Muscle, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Glycogen Storage Disease/physiopathology , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Protein-Arginine N-Methyltransferases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...