Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Eur J Nucl Med Mol Imaging ; 51(5): 1261-1267, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38095672

ABSTRACT

PURPOSE: Test the feasibility of an image-based method to identify taxane resistance in mouse bearing triple-negative breast cancer (TNBC) tumor xenografts. METHODS: Xenograft tumor-bearing mice from paclitaxel-sensitive and paclitaxel-resistant TNBC cells (MDA-MD-346) were generated by orthotopic injection into female NOD-SCID mice. When tumors reached 100-150 mm3, mice were scanned using [18F]choline PET/CT. Tumors were collected and sliced for autoradiography and immunofluorescence analysis. Quantitative data was analyzed accordingly. RESULTS: From fifteen mice scanned, five had taxane-sensitive cell line tumors of which two underwent taxol-based treatment. From the remaining 10 mice with taxane-resistant cell line tumors, four underwent taxol-based treatment. Only 13 mice had the tumor sample analyzed histologically. When normalized to the blood pool, both cell lines showed differences in metabolic uptake before and after treatment. CONCLUSIONS: Treated and untreated taxane-sensitive and taxane-resistant cell lines have different metabolic properties that could be leveraged before the start of chemotherapy.


Subject(s)
Positron Emission Tomography Computed Tomography , Triple Negative Breast Neoplasms , Humans , Female , Animals , Mice , Positron Emission Tomography Computed Tomography/methods , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Mice, SCID , Mice, Inbred NOD , Positron-Emission Tomography/methods , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Models, Animal , Drug Resistance , Xenograft Model Antitumor Assays
2.
Oncogene ; 42(21): 1693-1703, 2023 05.
Article in English | MEDLINE | ID: mdl-37020039

ABSTRACT

Predicting and treating recurrence in intermediate-risk prostate cancer patients remains a challenge despite having identified genomic instability [1] and hypoxia [2, 3] as risk factors. This underlies challenges in assigning the functional impact of these risk factors to mechanisms promoting prostate cancer progression. Here we show chronic hypoxia (CH), as observed in prostate tumours [4], leads to the adoption of an androgen-independent state in prostate cancer cells. Specifically, CH results in prostate cancer cells adopting transcriptional and metabolic alterations typical of castration-resistant prostate cancer cells. These changes include the increased expression of transmembrane transporters for the methionine cycle and related pathways leading to increased abundance of metabolites and expression of enzymes related to glycolysis. Targeting of the Glucose Transporter 1 (GLUT1) identified a dependency on glycolysis in androgen-independent cells. Overall, we identified a therapeutically targetable weakness in chronic hypoxia and androgen-independent prostate cancer. These findings may offer additional strategies for treatment development against hypoxic prostate cancer.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Androgens/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms/pathology , Prostate/pathology , Hypoxia/metabolism , Castration , Receptors, Androgen/genetics , Cell Line, Tumor
3.
Methods Mol Biol ; 2614: 313-348, 2023.
Article in English | MEDLINE | ID: mdl-36587133

ABSTRACT

Cancer cells within a tumor exhibit phenotypic plasticity that allows adaptation and survival in hostile tumor microenvironments. Reprogramming of epigenetic landscapes can support tumor progression within a specific microenvironment by influencing chromatin accessibility and modulating cell identity. The profiling of epigenetic landscapes within various tumor cell populations has significantly improved our understanding of tumor progression and plasticity. This protocol describes an integrated approach using chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) optimized to profile genome-wide post-translational modifications of histone tails in tumors. Essential tools amenable to ChIP-seq to isolate tumor cell populations of interest from the tumor microenvironment are also presented to provide a comprehensive approach to perform heterogeneous epigenetic landscape profiling of the tumor microenvironment.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Neoplasms , Humans , Chromatin Immunoprecipitation Sequencing/methods , Tumor Microenvironment/genetics , Histones/genetics , Histones/metabolism , Chromatin/genetics , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Epigenesis, Genetic
4.
Proc Natl Acad Sci U S A ; 119(36): e2203452119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037342

ABSTRACT

The contribution of deregulated chromatin architecture, including topologically associated domains (TADs), to cancer progression remains ambiguous. CCCTC-binding factor (CTCF) is a central regulator of higher-order chromatin structure that undergoes copy number loss in over half of all breast cancers, but the impact of this defect on epigenetic programming and chromatin architecture remains unclear. We find that under physiological conditions, CTCF organizes subTADs to limit the expression of oncogenic pathways, including phosphatidylinositol 3-kinase (PI3K) and cell adhesion networks. Loss of a single CTCF allele potentiates cell invasion through compromised chromatin insulation and a reorganization of chromatin architecture and histone programming that facilitates de novo promoter-enhancer contacts. However, this change in the higher-order chromatin landscape leads to a vulnerability to inhibitors of mTOR. These data support a model whereby subTAD reorganization drives both modification of histones at de novo enhancer-promoter contacts and transcriptional up-regulation of oncogenic transcriptional networks.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , CCCTC-Binding Factor/metabolism , Carcinogenesis/genetics , Chromatin/genetics , Chromatin/metabolism , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic
5.
Nat Chem Biol ; 18(8): 821-830, 2022 08.
Article in English | MEDLINE | ID: mdl-35578032

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with the worst prognosis and few effective therapies. Here we identified MS023, an inhibitor of type I protein arginine methyltransferases (PRMTs), which has antitumor growth activity in TNBC. Pathway analysis of TNBC cell lines indicates that the activation of interferon responses before and after MS023 treatment is a functional biomarker and determinant of response, and these observations extend to a panel of human-derived organoids. Inhibition of type I PRMT triggers an interferon response through the antiviral defense pathway with the induction of double-stranded RNA, which is derived, at least in part, from inverted repeat Alu elements. Together, our results represent a shift in understanding the antitumor mechanism of type I PRMT inhibitors and provide a rationale and biomarker approach for the clinical development of type I PRMT inhibitors.


Subject(s)
Protein-Arginine N-Methyltransferases , Triple Negative Breast Neoplasms , Biomarkers , Cell Line, Tumor , Humans , Interferons/therapeutic use , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
6.
Cancers (Basel) ; 13(18)2021 Sep 19.
Article in English | MEDLINE | ID: mdl-34572926

ABSTRACT

Breast cancer progression is characterized by changes in cellular metabolism that contribute to enhanced tumour growth and adaptation to microenvironmental stresses. Metabolic changes within breast tumours are still poorly understood and are not as yet exploited for therapeutic intervention, in part due to a high level of metabolic heterogeneity within tumours. The metabolic profiles of breast cancer cells are flexible, providing dynamic switches in metabolic states to accommodate nutrient and energy demands and further aggravating the challenges of targeting metabolic dependencies in cancer. In this review, we discuss the intrinsic and extrinsic factors that contribute to metabolic heterogeneity of breast tumours. Next, we examine how metabolic flexibility, which contributes to the metabolic heterogeneity of breast tumours, can alter epigenetic landscapes and increase a variety of pro-tumorigenic functions. Finally, we highlight the difficulties in pharmacologically targeting the metabolic adaptations of breast tumours and provide an overview of possible strategies to sensitize heterogeneous breast tumours to the targeting of metabolic vulnerabilities.

7.
JCI Insight ; 6(4)2021 02 22.
Article in English | MEDLINE | ID: mdl-33470989

ABSTRACT

Triple-negative breast cancers (TNBCs) lack effective targeted therapies, and cytotoxic chemotherapies remain the standard of care for this subtype. Owing to their increased genomic instability, poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) are being tested against TNBCs. In particular, clinical trials are now interrogating the efficacy of PARPi combined with chemotherapies. Intriguingly, while response rates are low, cohort of patients do respond to PARPi in combination with chemotherapies. Moreover, recent studies suggest that an increase in levels of ROS may sensitize cells to PARPi. This represents a therapeutic opportunity, as several chemotherapies, including doxorubicin, function in part by producing ROS. We previously demonstrated that the p66ShcA adaptor protein is variably expressed in TNBCs. We now show that, in response to therapy-induced stress, p66ShcA stimulated ROS production, which, in turn, potentiated the synergy of PARPi in combination with doxorubicin in TNBCs. This p66ShcA-induced sensitivity relied on the accumulation of oxidative damage in TNBCs, rather than genomic instability, to potentiate cell death. These findings suggest that increasing the expression of p66ShcA protein levels in TNBCs represents a rational approach to bolster the synergy between PARPi and doxorubicin.


Subject(s)
Antineoplastic Agents/pharmacology , Poly (ADP-Ribose) Polymerase-1/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Apoptosis , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Cell Survival , DNA Damage , Genomic Instability , Humans , MCF-7 Cells , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Src Homology 2 Domain-Containing, Transforming Protein 1 , Xenograft Model Antitumor Assays
8.
Cell Rep ; 32(12): 108170, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966787

ABSTRACT

The replication cycle and pathogenesis of the Plasmodium malarial parasite involves rapid expansion in red blood cells (RBCs), and variants of certain RBC-specific proteins protect against malaria in humans. In RBCs, bisphosphoglycerate mutase (BPGM) acts as a key allosteric regulator of hemoglobin/oxyhemoglobin. We demonstrate here that a loss-of-function mutation in the murine Bpgm (BpgmL166P) gene confers protection against both Plasmodium-induced cerebral malaria and blood-stage malaria. The malaria protection seen in BpgmL166P mutant mice is associated with reduced blood parasitemia levels, milder clinical symptoms, and increased survival. The protective effect of BpgmL166P involves a dual mechanism that enhances the host's stress erythroid response to Plasmodium-driven RBC loss and simultaneously alters the intracellular milieu of the RBCs, including increased oxyhemoglobin and reduced energy metabolism, reducing Plasmodium maturation, and replication. Overall, our study highlights the importance of BPGM as a regulator of hemoglobin/oxyhemoglobin in malaria pathogenesis and suggests a new potential malaria therapeutic target.


Subject(s)
Anemia/etiology , Anemia/prevention & control , Bisphosphoglycerate Mutase/deficiency , Malaria, Cerebral/enzymology , Malaria, Cerebral/prevention & control , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Bisphosphoglycerate Mutase/chemistry , Bisphosphoglycerate Mutase/genetics , Bisphosphoglycerate Mutase/metabolism , Enzyme Stability , Erythrocytes/enzymology , Erythrocytes/parasitology , Erythropoiesis , Extracellular Matrix/metabolism , Female , HEK293 Cells , Humans , Malaria, Cerebral/complications , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Parasites/growth & development , Plasmodium/growth & development , Polycythemia
9.
Nat Commun ; 11(1): 4205, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32826891

ABSTRACT

Triple negative breast cancer (TNBC) is a deadly form of breast cancer due to the development of resistance to chemotherapy affecting over 30% of patients. New therapeutics and companion biomarkers are urgently needed. Recognizing the elevated expression of glucose transporter 1 (GLUT1, encoded by SLC2A1) and associated metabolic dependencies in TNBC, we investigated the vulnerability of TNBC cell lines and patient-derived samples to GLUT1 inhibition. We report that genetic or pharmacological inhibition of GLUT1 with BAY-876 impairs the growth of a subset of TNBC cells displaying high glycolytic and lower oxidative phosphorylation (OXPHOS) rates. Pathway enrichment analysis of gene expression data suggests that the functionality of the E2F pathway may reflect to some extent OXPHOS activity. Furthermore, the protein levels of retinoblastoma tumor suppressor (RB1) strongly correlate with the degree of sensitivity to GLUT1 inhibition in TNBC, where RB1-negative cells are insensitive to GLUT1 inhibition. Collectively, our results highlight a strong and targetable RB1-GLUT1 metabolic axis in TNBC and warrant clinical evaluation of GLUT1 inhibition in TNBC patients stratified according to RB1 protein expression levels.


Subject(s)
Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/metabolism , Retinoblastoma Binding Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Biomarkers, Tumor , Breast Neoplasms/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucose Transporter Type 1/genetics , Humans , Mice , Oxidative Phosphorylation , Proteomics , Pyrazoles/pharmacology , Pyridines/pharmacology , Quinolines , RNA, Messenger/metabolism , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
10.
Cancer Discov ; 10(9): 1312-1329, 2020 09.
Article in English | MEDLINE | ID: mdl-32546577

ABSTRACT

Tumor progression upon treatment arises from preexisting resistant cancer cells and/or adaptation of persister cancer cells committing to an expansion phase. Here, we show that evasion from viral mimicry response allows the growth of taxane-resistant triple-negative breast cancer (TNBC). This is enabled by an epigenetic state adapted to taxane-induced metabolic stress, where DNA hypomethylation over loci enriched in transposable elements (TE) is compensated by large chromatin domains of H3K27me3 to warrant TE repression. This epigenetic state creates a vulnerability to epigenetic therapy against EZH2, the H3K27me3 methyltransferase, which alleviates TE repression in taxane-resistant TNBC, leading to double-stranded RNA production and growth inhibition through viral mimicry response. Collectively, our results illustrate how epigenetic states over TEs promote cancer progression under treatment and can inform about vulnerabilities to epigenetic therapy. SIGNIFICANCE: Drug-resistant cancer cells represent a major barrier to remission for patients with cancer. Here we show that drug-induced metabolic perturbation and epigenetic states enable evasion from the viral mimicry response induced by chemotherapy in TNBC. These epigenetic states define a vulnerability to epigenetic therapy using EZH2 inhibitors in taxane-resistant TNBC.See related commentary by Janin and Esteller, p. 1258.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/immunology , Molecular Mimicry/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Escape/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chromatin Immunoprecipitation Sequencing , DNA Methylation/drug effects , DNA Methylation/immunology , DNA Transposable Elements/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic/drug effects , Female , Humans , Mice , Molecular Mimicry/genetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
11.
Cell ; 181(6): 1329-1345.e24, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32445698

ABSTRACT

Posterior fossa A (PFA) ependymomas are lethal malignancies of the hindbrain in infants and toddlers. Lacking highly recurrent somatic mutations, PFA ependymomas are proposed to be epigenetically driven tumors for which model systems are lacking. Here we demonstrate that PFA ependymomas are maintained under hypoxia, associated with restricted availability of specific metabolites to diminish histone methylation, and increase histone demethylation and acetylation at histone 3 lysine 27 (H3K27). PFA ependymomas initiate from a cell lineage in the first trimester of human development that resides in restricted oxygen. Unlike other ependymomas, transient exposure of PFA cells to ambient oxygen induces irreversible cellular toxicity. PFA tumors exhibit a low basal level of H3K27me3, and, paradoxically, inhibition of H3K27 methylation specifically disrupts PFA tumor growth. Targeting metabolism and/or the epigenome presents a unique opportunity for rational therapy for infants with PFA ependymoma.


Subject(s)
Ependymoma/genetics , Ependymoma/metabolism , Epigenome/genetics , Infratentorial Neoplasms/genetics , Infratentorial Neoplasms/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line , Cell Proliferation/genetics , DNA Methylation/genetics , Epigenomics/methods , Histones/genetics , Histones/metabolism , Humans , Infant , Lysine/genetics , Lysine/metabolism , Male , Mice, Inbred C57BL , Mutation/genetics
12.
Nat Commun ; 10(1): 1915, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015424

ABSTRACT

Bromodomains (BRDs) are conserved protein interaction modules which recognize (read) acetyl-lysine modifications, however their role(s) in regulating cellular states and their potential as targets for the development of targeted treatment strategies is poorly understood. Here we present a set of 25 chemical probes, selective small molecule inhibitors, covering 29 human bromodomain targets. We comprehensively evaluate the selectivity of this probe-set using BROMOscan and demonstrate the utility of the set identifying roles of BRDs in cellular processes and potential translational applications. For instance, we discovered crosstalk between histone acetylation and the glycolytic pathway resulting in a vulnerability of breast cancer cell lines under conditions of glucose deprivation or GLUT1 inhibition to inhibition of BRPF2/3 BRDs. This chemical probe-set will serve as a resource for future applications in the discovery of new physiological roles of bromodomain proteins in normal and disease states, and as a toolset for bromodomain target validation.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/pharmacology , Acetylation , Amino Acid Sequence , Antineoplastic Agents/chemistry , Cell Line, Tumor , Epigenesis, Genetic , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glucose/deficiency , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Glycolysis/genetics , High-Throughput Screening Assays , Histone Acetyltransferases , Histone Chaperones , Histones/genetics , Histones/metabolism , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship
13.
J Cell Biol ; 217(8): 2951-2974, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29921600

ABSTRACT

The mammary epithelium depends on specific lineages and their stem and progenitor function to accommodate hormone-triggered physiological demands in the adult female. Perturbations of these lineages underpin breast cancer risk, yet our understanding of normal mammary cell composition is incomplete. Here, we build a multimodal resource for the adult gland through comprehensive profiling of primary cell epigenomes, transcriptomes, and proteomes. We define systems-level relationships between chromatin-DNA-RNA-protein states, identify lineage-specific DNA methylation of transcription factor binding sites, and pinpoint proteins underlying progesterone responsiveness. Comparative proteomics of estrogen and progesterone receptor-positive and -negative cell populations, extensive target validation, and drug testing lead to discovery of stem and progenitor cell vulnerabilities. Top epigenetic drugs exert cytostatic effects; prevent adult mammary cell expansion, clonogenicity, and mammopoiesis; and deplete stem cell frequency. Select drugs also abrogate human breast progenitor cell activity in normal and high-risk patient samples. This integrative computational and functional study provides fundamental insight into mammary lineage and stem cell biology.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Lineage , DNA Methylation , DNA, Neoplasm/metabolism , Epigenesis, Genetic/drug effects , Epigenomics , Humans , Mice , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Progesterone/pharmacology , Proteome , RNA, Neoplasm/metabolism , Risk Factors , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
14.
Cell Rep ; 16(7): 1829-37, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27498878

ABSTRACT

Pro-inflammatory signals provided by the microenvironment are critical to activate dendritic cells (DCs), components of the innate immune system that shape both innate and adaptive immunity. However, to prevent inappropriate immune activation, mechanisms must be in place to restrain DC activation to ensure DCs are activated only once sufficient stimuli have been received. Here, we report that DC activation and immunogenicity are regulated by the transcriptional repressor Polycomb group factor 6 (PCGF6). Pcgf6 is rapidly downregulated upon stimulation, and this downregulation is necessary to permit full DC activation. Silencing PCGF6 expression enhanced both spontaneous and stimulated DC activation. We show that PCGF6 associates with the H3K4me3 demethylase JARID1c, and together, they negatively regulate H3K4me3 levels in DCs. Our results identify two key regulators, PCGF6 and JARID1c that temper DC activation and implicate active transcriptional silencing via histone demethylation as a previously unappreciated mechanism for regulating DC activation and quiescence.


Subject(s)
Dendritic Cells/immunology , Histones/genetics , Oxidoreductases, N-Demethylating/genetics , Polycomb Repressive Complex 1/genetics , Repressor Proteins/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Chromatin/chemistry , Chromatin/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Female , Gene Expression Regulation , Histone Demethylases , Histones/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidoreductases, N-Demethylating/immunology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/immunology , Signal Transduction , Transcription, Genetic
15.
Nat Commun ; 7: 12156, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27402251

ABSTRACT

Despite the initial benefits of treating HER2-amplified breast cancer patients with the tyrosine kinase inhibitor lapatinib, resistance inevitably develops. Here we report that lapatinib induces the degradation of the nuclear receptor ERRα, a master regulator of cellular metabolism, and that the expression of ERRα is restored in lapatinib-resistant breast cancer cells through reactivation of mTOR signalling. Re-expression of ERRα in resistant cells triggers metabolic adaptations favouring mitochondrial energy metabolism through increased glutamine metabolism, as well as ROS detoxification required for cell survival under therapeutic stress conditions. An ERRα inverse agonist counteracts these metabolic adaptations and overcomes lapatinib resistance in a HER2-induced mammary tumour mouse model. This work reveals a molecular mechanism by which ERRα-induced metabolic reprogramming promotes survival of lapatinib-resistant cancer cells and demonstrates the potential of ERRα inhibition as an effective adjuvant therapy in poor outcome HER2-positive breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Mammary Neoplasms, Experimental/drug therapy , Quinazolines/therapeutic use , Receptors, Estrogen/genetics , Animals , Breast Neoplasms/metabolism , Cell Survival , Humans , Lapatinib , MCF-7 Cells , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse , Mice , Receptor, ErbB-2/metabolism , Retroviridae Infections , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Virus Infections , ERRalpha Estrogen-Related Receptor
16.
Oncotarget ; 7(31): 49611-49622, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27391063

ABSTRACT

microRNA-34A is a critical component of the p53 network and expression of miR- 34A is down-regulated by promoter hypermethylation or focal deletions in numerous human cancers. Although miR-34A deregulation may be an important driver in cancer, the endogenous role of this microRNA in cellular homeostasis is not well characterized. To address this knowledge gap, we aimed to determine the transcriptional landscape of the miR-34A-p53 axis in non-transformed cells. Using primary skin-derived fibroblast cell lines from patients who developed childhood cancers, and who harbor either germline TP53 mutations or are TP53 wild type, we sought to characterize the transcriptional response to miR-34A modulation. Through transcriptome-wide RNA-Sequencing, we show for the first time that in human non- transformed cells harboring TP53 mutations, miR-34A functions in a noncanonical manner to influence noncoding RNA networks, including RNA components of the minor (U12) spliceosome, as well as TP53-dependent and independent epigenetic pathways. miR- 34A-regulated transcripts include known cell cycle mediators and abrogation of miR-34A leads to a TP53-dependent increase in the fraction of cells in G2/M. Collectively, these results provide a framework for understanding the endogenous role of the miR-34A signaling axis and identify novel transcripts and pathways regulated by the essential miR-34A-p53 tumor suppressor network.


Subject(s)
Genes, Tumor Suppressor , MicroRNAs/metabolism , Transcriptome , Tumor Suppressor Protein p53/genetics , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Child , Child, Preschool , DNA Mutational Analysis , Epigenesis, Genetic , Gene Expression Profiling , Homeostasis , Humans , Infant , MicroRNAs/genetics , Neoplasms/genetics , Promoter Regions, Genetic , Sequence Analysis, RNA
17.
Mol Cell Biol ; 34(23): 4232-43, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25246633

ABSTRACT

The tyrosine kinase receptor ERBB2 is required for normal development of the heart and is a potent oncogene in breast epithelium. Trastuzumab, a monoclonal antibody targeting ERBB2, improves the survival of breast cancer patients, but cardiac dysfunction is a major side effect of the drug. The molecular mechanisms underlying how ERBB2 regulates cardiac function and why trastuzumab is cardiotoxic remain poorly understood. We show here that ERBB2 hypomorphic mice develop cardiac dysfunction that mimics the side effects observed in patients treated with trastuzumab. We demonstrate that this phenotype is related to the critical role played by ERBB2 in cardiac homeostasis and physiological hypertrophy. Importantly, genetic and therapeutic reduction of ERBB2 activity in mice, as well as ablation of ERBB2 signaling by trastuzumab or siRNAs in human cardiomyocytes, led to the identification of an impaired E2F-1-dependent genetic program critical for the cardiac adaptive stress response. These findings demonstrate the existence of a previously unknown mechanistic link between ERBB2 and E2F-1 transcriptional activity in heart physiology and trastuzumab-induced cardiac dysfunction.


Subject(s)
Adaptation, Physiological , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Cardiomegaly/genetics , E2F1 Transcription Factor/biosynthesis , Myocytes, Cardiac/drug effects , Receptor, ErbB-2/genetics , Stress, Physiological , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Cells, Cultured , Doxorubicin/adverse effects , Doxorubicin/pharmacology , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Echocardiography , Fibrosis , Gene Expression Profiling , Gene Knock-In Techniques , Heart/growth & development , Humans , Mice , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/immunology , Signal Transduction/genetics , Stroke Volume/genetics , Trastuzumab
18.
Mol Cancer Res ; 12(2): 184-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24379448

ABSTRACT

UNLABELLED: The 20q13 chromosomal region has been previously identified as the hereditary prostate cancer genetic-susceptibility locus on chromosome 20 (HPC20). In this study, the 20q13 region was shown to be frequently co-amplified with the androgen receptor (AR) in metastatic prostate cancer. Furthermore, the AR signaling axis, which plays an essential role in the pathogenesis of prostate cancer, was demonstrated to be central to the regulation of the 20q13 common amplified region (CAR). High-resolution mapping analyses revealed hot spots of AR recruitment to response elements in the vicinity of most genes located on the 20q13 CAR. Moreover, amplification of AR significantly co-occurred with CAR amplification on 20q13 and it was confirmed that the majority of AR-bound genes on the 20q13 CAR were indeed regulated by androgens. These data reveal that amplification of the AR is tightly linked to amplification of the AR-regulated CAR region on 20q13. These results suggest that the cross-talk between gene amplification and gene transcription is an important step in the development of castration-resistant metastatic disease. IMPLICATIONS: These novel results are a noteworthy example of the cross-talk between gene amplification and gene transcription in the development of advanced prostate cancer.


Subject(s)
Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 20 , Gene Amplification , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Loci , Genetic Predisposition to Disease , Humans , Male , Receptors, Androgen/metabolism
19.
Cancer Metab ; 1(1): 22, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24304688

ABSTRACT

BACKGROUND: Glutamine metabolism is a central metabolic pathway in cancer. Recently, reductive carboxylation of glutamine for lipogenesis has been shown to constitute a key anabolic route in cancer cells. However, little is known regarding central regulators of the various glutamine metabolic pathways in cancer cells. METHODS: The impact of PGC-1α and ERRα on glutamine enzyme expression was assessed in ERBB2+ breast cancer cell lines with quantitative RT-PCR, chromatin immunoprecipitation, and immunoblotting experiments. Glutamine flux was quantified using 13C-labeled glutamine and GC/MS analyses. Functional assays for lipogenesis were performed using 14C-labeled glutamine. The expression of glutamine metabolism genes in breast cancer patients was determined by bioinformatics analyses using The Cancer Genome Atlas. RESULTS: We show that the transcriptional coactivator PGC-1α, along with the transcription factor ERRα, is a positive regulator of the expression of glutamine metabolism genes in ERBB2+ breast cancer. Indeed, ERBB2+ breast cancer cells with increased expression of PGC-1α display elevated expression of glutamine metabolism genes. Furthermore, ERBB2+ breast cancer cells with reduced expression of PGC-1α or when treated with C29, a pharmacological inhibitor of ERRα, exhibit diminished expression of glutamine metabolism genes. The biological relevance of the control of glutamine metabolism genes by the PGC-1α/ERRα axis is demonstrated by consequent regulation of glutamine flux through the citric acid cycle. PGC-1α and ERRα regulate both the canonical citric acid cycle (forward) and the reductive carboxylation (reverse) fluxes; the latter can be used to support de novo lipogenesis reactions, most notably in hypoxic conditions. Importantly, murine and human ERBB2+ cells lines display a significant dependence on glutamine availability for their growth. Finally, we show that PGC-1α expression is positively correlated with that of the glutamine pathway in ERBB2+ breast cancer patients, and high expression of this pathway is associated with reduced patient survival. CONCLUSIONS: These data reveal that the PGC-1α/ERRα axis is a central regulator of glutamine metabolism in ERBB2+ breast cancer. This novel regulatory link, as well as the marked reduction in patient survival time associated with increased glutamine pathway gene expression, suggests that targeting glutamine metabolism may have therapeutic potential in the treatment of ERBB2+ breast cancer.

20.
Cancer Res ; 73(14): 4474-87, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23720052

ABSTRACT

Although ERBB2 amplification and overexpression is correlated with poor outcome in breast cancer, the molecular mechanisms underlying the aggressive nature of these tumors has not been fully elucidated. To investigate this further, we have used a transgenic mouse model of ErbB2-driven tumor progression (ErbB2(KI) model) that recapitulates clinically relevant events, including selective amplification of the core erbB2 amplicon. By comparing the transcriptional profiles of ErbB2(KI) mammary tumors and human ERBB2-positive breast cancers, we show that ErbB2(KI) tumors possess molecular features of the basal subtype of ERBB2-positive human breast cancer, including activation of canonical ß-catenin signaling. Inhibition of ß-catenin-dependent signaling in ErbB2(KI)-derived tumor cells using RNA interference impaired tumor initiation and metastasis. Furthermore, treatment of ErbB2(KI) or human ERBB2-overexpressing tumor cells with a selective ß-catenin/CBP inhibitor significantly decreased proliferation and ErbB2 expression. Collectively, our data indicate that ERBB2-mediated breast cancer progression requires ß-catenin signaling and can be therapeutically targeted by selective ß-catenin/CBP inhibitors.


Subject(s)
Breast Neoplasms/pathology , Mammary Neoplasms, Experimental/pathology , Receptor, ErbB-2/metabolism , beta Catenin/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Progression , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Receptor, ErbB-2/genetics , Signal Transduction , Transcription, Genetic , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...