Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
iScience ; 24(10): 103212, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34755086

ABSTRACT

Activity of liver x receptor (LXR), the homeostatic regulator of cholesterol metabolism, is elevated in triple-negative breast cancer (BCa) relative to other BCa subtypes, driving drug resistance and metastatic gene signatures. The loci encoding LXRα and LXRß produce multiple alternatively spliced proteins, but the true range of variants and their relevance to cancer remain poorly defined. Here, we report seven LXR splice variants, three of which have not previously been reported and five that were prognostic for disease-free survival. Expression of full-length LXRα splice variants was associated with poor prognosis, consistent with a role as an oncogenic driver of triple-negative tumor pathophysiology. Contrary to this was the observation that high expression of truncated LXRα splice variants or any LXRß splice variant was associated with longer survival. These findings indicate that LXR isoform abundance is an important aspect of understanding the link between dysregulated cholesterol metabolism and cancer pathophysiology.

2.
Oncogene ; 40(16): 2872-2883, 2021 04.
Article in English | MEDLINE | ID: mdl-33742124

ABSTRACT

Triple negative breast cancer (TNBC) is challenging to treat successfully because targeted therapies do not exist. Instead, systemic therapy is typically restricted to cytotoxic chemotherapy, which fails more often in patients with elevated circulating cholesterol. Liver x receptors are ligand-dependent transcription factors that are homeostatic regulators of cholesterol, and are linked to regulation of broad-affinity xenobiotic transporter activity in non-tumor tissues. We show that LXR ligands confer chemotherapy resistance in TNBC cell lines and xenografts, and that LXRalpha is necessary and sufficient to mediate this resistance. Furthermore, in TNBC patients who had cancer recurrences, LXRalpha and ligands were independent markers of poor prognosis and correlated with P-glycoprotein expression. However, in patients who survived their disease, LXRalpha signaling and P-glycoprotein were decoupled. These data reveal a novel chemotherapy resistance mechanism in this poor prognosis subtype of breast cancer. We conclude that systemic chemotherapy failure in some TNBC patients is caused by co-opting the LXRalpha:P-glycoprotein axis, a pathway highly targetable by therapies that are already used for prevention and treatment of other diseases.


Subject(s)
Hydroxycholesterols/metabolism , Liver X Receptors/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Epirubicin/pharmacology , Female , Gene Expression , Humans , Liver X Receptors/agonists , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
3.
Nutrients ; 11(11)2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31683867

ABSTRACT

Interventions that alter cholesterol have differential impacts on hormone receptor positive- and negative-breast cancer risk and prognosis. This implies differential regulation or response to cholesterol within different breast cancer subtypes. We evaluated differences in side-chain hydroxycholesterol and liver X nuclear receptor signalling between Oestrogen Receptor (ER)-positive and ER-negative breast cancers and cell lines. Cell line models of ER-positive and ER-negative disease were treated with Liver X Receptor (LXR) ligands and transcriptional activity assessed using luciferase reporters, qPCR and MTT. Publicly available datasets were mined to identify differences between ER-negative and ER-positive tumours and siRNA was used to suppress candidate regulators. Compared to ER-positive breast cancer, ER-negative breast cancer cells were highly responsive to LXR agonists. In primary disease and cell lines LXRA expression was strongly correlated with its target genes in ER-negative but not ER-positive disease. Expression of LXR's corepressors (NCOR1, NCOR2 and LCOR) was significantly higher in ER-positive disease relative to ER-negative, and their knock-down equalized sensitivity to ligand between subtypes in reporter, gene expression and viability assays. Our data support further evaluation of dietary and pharmacological targeting of cholesterol metabolism as an adjunct to existing therapies for ER-negative and ER-positive breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Cholesterol/metabolism , Gene Expression Regulation, Neoplastic , Liver X Receptors/metabolism , Receptors, Estrogen/metabolism , Repressor Proteins/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Prognosis , RNA, Small Interfering , Signal Transduction , Transcription, Genetic
4.
Int J Mol Sci ; 20(13)2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31269628

ABSTRACT

Low fruit and vegetable consumption and high saturated fat consumption causes elevated circulating cholesterol and are breast cancer risk factors. During cholesterol metabolism, oxysterols form that bind and activate the liver X receptors (LXRs). Oxysterols halt breast cancer cell proliferation but enhance metastatic colonization, indicating tumour suppressing and promoting roles. Phytosterols and phytostanols in plants, like cholesterol in mammals, are essential components of the plasma membrane and biochemical precursors, and in human cells can alter LXR transcriptional activity. Here, a panel of breast cancer cell lines were treated with four dietary plant sterols and a stanol, alone or in combination with oxysterols. LXR activation and repression were measured by gene expression and LXR-luciferase reporter assays. Oxysterols activated LXR in all cell lines, but surprisingly phytosterols failed to modulate LXR activity. However, phytosterols significantly inhibited the ability of oxysterols to drive LXR transcription. These data support a role for phytosterols in modulating cancer cell behaviour via LXR, and therefore suggest merit in accurate dietary recordings of these molecules in cancer patients during treatment and perhaps supplementation to benefit recovery.


Subject(s)
Breast Neoplasms/genetics , Liver X Receptors/genetics , Oxysterols/metabolism , Phytosterols/pharmacology , Transcriptional Activation/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Liver X Receptors/metabolism
5.
Methods Mol Biol ; 1951: 15-32, 2019.
Article in English | MEDLINE | ID: mdl-30825141

ABSTRACT

Nuclear receptors (NRs) are ligand-activated transcription factors. Class 2 NRs, such as the liver X receptor (LXR)α and (LXR)ß, are typically retained in the nucleus bound to the DNA in both the presence and absence of ligand. Upon binding ligands including hydroxylated cholesterol, LXR releases corepressor proteins in exchange for coactivators resulting in target gene transcription. Activity of the LXRs therefore depends on a combination of the local ligand concentration(s) and cofactor expression, which itself is a function of cell and tissue type, mutation load, and epigenetic regulation. Cross talk with other transcription factors or signaling pathways can also alter LXR activity. The role that LXR plays in both normal physiology and disease progression is becoming increasingly apparent, and a better understanding of how and when LXR is activated or repressed is pressing biological and clinical questions.The complexity of LXR regulation makes identifying novel ligands and determining LXR activity in new cell types challenging. Generating cell lines that contain a stably integrated luciferase reporter gene with an upstream LXR-dependent promoter provides a quick, cheap, robust, efficient, and high-throughput solution to identify novel ligands and assess ligand activity in new cell types. Transplant of these stable cell culture cell lines as xenografts allows reporter activation to be assessed in vivo. Here we describe the generation of stable LXR reporter cell lines, how to confirm transgene insertion and select single cell clones, as well a method to assess transgene activity in vitro.


Subject(s)
Gene Expression Regulation/drug effects , Genes, Reporter , Liver X Receptors/genetics , Oxysterols/pharmacology , Data Analysis , Gene Dosage , Genetic Vectors/genetics , Humans , Ligands , Liver X Receptors/metabolism , Transduction, Genetic
6.
J Steroid Biochem Mol Biol ; 192: 105309, 2019 09.
Article in English | MEDLINE | ID: mdl-30779932

ABSTRACT

Oxysterols can contribute to proliferation of breast cancer through activation of the Estrogen Receptors, and to metastasis through activation of the Liver X Receptors. Endogenous levels of both esterified and free sidechain-hydroxylated oxysterols were examined in breast cancer tumours from Estrogen Receptor positive and negative breast tumours, using a novel fast liquid chromatography tandem mass spectrometry method. Multiple aliquots of five milligram samples of 22 tumours were analysed for oxysterol content to assess intra- and inter-tumour variation. Derivatization was performed with Girard T reagent (with and without alkaline hydrolysis) and sample clean-up was performed using a robust automatic on-line column switching system ("AFFL"). Oxysterols were separated isocratically on a 2.1 mm inner diameter column packed with ACE SuperPhenylHexyl core shell particles using a mobile phase consisting of 0.1% formic acid in H2O/methanol/acetonitrile (57/10/33, v/v/v) followed by a wash out step (0.1% formic acid in methanol/acetonitrile, 50/50, v/v). The total analysis time, including sample clean-up and column reconditioning, was 8 min (80% time reduction compared to other on-line systems). Analysis revealed large intra-tumour variations of sidechain oxysterols, resulting in no significant differences in endogenous oxysterols levels between Estrogen Receptor positive and Estrogen Receptor negative breast cancers. However, a correlation between esterified and free 27-hydroxycholesterol was observed. The same correlation was not observed for 24S-hydroxycholesterol or 25-hydroxycholesterol. The oxysterol heterogeneity of tumour tissue is a critical factor when assessing the role of these lipids in cancer.


Subject(s)
Breast Neoplasms/metabolism , Chromatography, Liquid/methods , Oxysterols/analysis , Oxysterols/chemistry , Tandem Mass Spectrometry/methods , Breast Neoplasms/pathology , Female , Humans , Hydroxycholesterols/metabolism
7.
Biochim Biophys Acta Gene Regul Mech ; 1861(11): 996-1006, 2018 11.
Article in English | MEDLINE | ID: mdl-30343695

ABSTRACT

MicroRNAs and RNA-binding proteins exert regulation on >60% of coding genes, yet interplay between them is little studied. Canonical microRNA binding occurs by base-pairing of microRNA 3'-ends to complementary "seed regions" in mRNA 3'UTRs, resulting in translational repression. Similarly, regulatory RNA-binding proteins bind to mRNAs, modifying stability or translation. We investigated post-transcriptional regulation acting on the xenobiotic pump ABCB1/P-glycoprotein, which is implicated in cancer therapy resistance. We characterised the ABCB1 UTRs in primary breast cancer cells and identified UTR sequences that responded to miR-19b despite lacking a canonical binding site. Sequences did, however, contain consensus sites for the RNA-binding protein HuR. We demonstrated that a tripartite complex of HuR, miR-19b and UTR directs repression of ABCB1/P-glycoprotein expression, with HuR essential for non-canonical miR-19b binding thereby controlling chemosensitivity of breast cancer cells. This exemplifies a new cooperative model between RNA-binding proteins and microRNAs to expand the repertoire of mRNAs that can be regulated. This study suggests a novel therapeutic target to impair P-glycoprotein mediated drug efflux, and also indicates that current microRNA binding predictions that rely on seed regions alone may be too conservative.


Subject(s)
Breast Neoplasms/metabolism , ELAV-Like Protein 1/metabolism , MicroRNAs/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antibiotics, Antineoplastic/pharmacology , Breast/metabolism , Cell Line , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Humans , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL