Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Proc Natl Acad Sci U S A ; 112(37): 11583-8, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26324937

ABSTRACT

The tumor protein 53 (TP53) tumor suppressor gene is the most frequently somatically altered gene in human cancers. Here we show expression of N-Myc down-regulated gene 1 (NDRG1) is induced by p53 during physiologic low proliferative states, and mediates centrosome homeostasis, thus maintaining genome stability. When placed in physiologic low-proliferating conditions, human TP53 null cells fail to increase expression of NDRG1 compared with isogenic wild-type controls and TP53 R248W knockin cells. Overexpression and RNA interference studies demonstrate that NDRG1 regulates centrosome number and amplification. Mechanistically, NDRG1 physically associates with ƎĀ³-tubulin, a key component of the centrosome, with reduced association in p53 null cells. Strikingly, TP53 homozygous loss was mutually exclusive of NDRG1 overexpression in over 96% of human cancers, supporting the broad applicability of these results. Our study elucidates a mechanism of how TP53 loss leads to abnormal centrosome numbers and genomic instability mediated by NDRG1.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/ultrastructure , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Aneuploidy , Animals , Breast/metabolism , Cell Line , Cell Proliferation , Centrosome/metabolism , Female , Genome , Heterozygote , Homeostasis , Homozygote , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Neoplasms/pathology , Phenotype , RNA Interference , Tubulin/metabolism
2.
Proc Natl Acad Sci U S A ; 111(49): 17606-11, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25422431

ABSTRACT

Tamoxifen is effective for treating estrogen receptor-alpha (ER) positive breast cancers. However, few molecular mediators of tamoxifen resistance have been elucidated. Here we describe a previously unidentified gene, MACROD2 that confers tamoxifen resistance and estrogen independent growth. We found MACROD2 is amplified and overexpressed in metastatic tamoxifen-resistant tumors. Transgene overexpression of MACROD2 in breast cancer cell lines results in tamoxifen resistance, whereas RNAi-mediated gene knock down reverses this phenotype. MACROD2 overexpression also leads to estrogen independent growth in xenograft assays. Mechanistically, MACROD2 increases p300 binding to estrogen response elements in a subset of ER regulated genes. Primary breast cancers and matched metastases demonstrate MACROD2 expression can change with disease evolution, and increased expression and amplification of MACROD2 in primary tumors is associated with worse overall survival. These studies establish MACROD2 as a key mediator of estrogen independent growth and tamoxifen resistance, as well as a potential novel target for diagnostics and therapy.


Subject(s)
Breast Neoplasms/metabolism , DNA Repair Enzymes/metabolism , Drug Resistance, Neoplasm , Estrogens/metabolism , Hydrolases/metabolism , Tamoxifen/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation , Epigenesis, Genetic , Female , Gene Deletion , Gene Dosage , Humans , Molecular Sequence Data , Neoplasm Transplantation , Phenotype , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Transgenes , Treatment Outcome
3.
Proteomics ; 15(2-3): 318-26, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25367220

ABSTRACT

The PIK3CA gene encodes for the p110 alpha isoform of PI3 kinase and is one of the most frequently mutated oncogenes in human cancers. However, the mechanisms by which PIK3CA mutations activate cell signaling are not fully understood. Here we used a phosphoproteomic approach to compare differential phosphorylation patterns between human breast epithelial cells and two isogenic somatic cell knock in derivatives, each harboring a distinct PIK3CA mutation. We demonstrated differential phosphorylation patterns between isogenic cell lines containing a PIK3CA helical domain mutation (E545K) compared to cells with a PIK3CA kinase domain mutation (H1047R). In particular, the receptor tyrosine kinase, HER3, showed increased phosphorylation at tyrosine 1328 in H1047R cells versus E545K cells. Genetic studies using shRNA demonstrated that H1047R cells have a profound decrease in growth factor independent proliferation upon HER3 knock down, but this effect was attenuated in E545K cells. In addition, HER3 knock down led to reductions in both PI3 kinase and MAP kinase pathway activation in H1047R cells, but in E545K cells only PI3 kinase pathway diminution was observed. These studies demonstrate the power of using paired isogenic cell lines for proteomic analysis to gain new insights into oncogenic signal transduction pathways.


Subject(s)
Breast Neoplasms/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteomics , Receptor, ErbB-3/metabolism , Signal Transduction , Breast Neoplasms/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Protein Structure, Tertiary , RNA Interference , Receptor, ErbB-3/genetics
4.
Proc Natl Acad Sci U S A ; 108(43): 17773-8, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21987798

ABSTRACT

Biallelic inactivation of cancer susceptibility gene BRCA1 leads to breast and ovarian carcinogenesis. Paradoxically, BRCA1 deficiency in mice results in early embryonic lethality, and similarly, lack of BRCA1 in human cells is thought to result in cellular lethality in view of BRCA1's essential function. To survive homozygous BRCA1 inactivation during tumorigenesis, precancerous cells must accumulate additional genetic alterations, such as p53 mutations, but this requirement for an extra genetic "hit" contradicts the two-hit theory for the accelerated carcinogenesis associated with familial cancer syndromes. Here, we show that heterozygous BRCA1 inactivation results in genomic instability in nontumorigenic human breast epithelial cells in vitro and in vivo. Using somatic cell gene targeting, we demonstrated that a heterozygous BRCA1 185delAG mutation confers impaired homology-mediated DNA repair and hypersensitivity to genotoxic stress. Heterozygous mutant BRCA1 cell clones also showed a higher degree of gene copy number loss and loss of heterozygosity in SNP array analyses. In BRCA1 heterozygous clones and nontumorigenic breast epithelial tissues from BRCA mutation carriers, FISH revealed elevated genomic instability when compared with their respective controls. Thus, BRCA1 haploinsufficiency may accelerate hereditary breast carcinogenesis by facilitating additional genetic alterations.


Subject(s)
Breast/cytology , Epithelial Cells/physiology , Genes, BRCA1 , Genomic Instability/genetics , Haploinsufficiency/genetics , Female , Gene Silencing , Genomic Instability/physiology , Heterozygote , Humans , In Situ Hybridization, Fluorescence , Polymorphism, Single Nucleotide , Sequence Deletion/genetics
5.
Fetal Diagn Ther ; 36(4): 282-6, 2014.
Article in English | MEDLINE | ID: mdl-25228026

ABSTRACT

OBJECTIVE: To develop a microarray-based method for noninvasive prenatal testing (NIPT) and compare it with next-generation sequencing. METHODS: Maternal plasma from 878 pregnant women, including 187 trisomy cases (18 trisomy 13, 37 trisomy 18, 132 trisomy 21), was evaluated for trisomy risk. Targeted chromosomes were analyzed using Digital Analysis of Selected Regions (DANSR™) assays. DANSR products were subsequently divided between two DNA quantification methods: microarrays and next-generation sequencing. For both microarray and sequencing methodologies, the Fetal-Fraction Optimized Risk of Trisomy Evaluation (FORTE™) algorithm was used to determine trisomy risk, assay variability across samples, and compute fetal fraction variability within samples. RESULTS: NIPT using microarrays provided faster and more accurate cell-free DNA (cfDNA) measurements than sequencing. The assay variability, a measure of variance of chromosomal cfDNA counts, was lower for microarrays than for sequencing, 0.051 versus 0.099 (p < 0.0001). Analysis time using microarrays was faster, 7.5 versus 56 h for sequencing. Additionally, fetal fraction precision was improved 1.6-fold by assaying more polymorphic sites with microarrays (p < 0.0001). Microarrays correctly classified all trisomy and nontrisomy cases. CONCLUSIONS: NIPT using microarrays delivers more accurate cfDNA analysis than next-generation sequencing and can be performed in less time.


Subject(s)
Oligonucleotide Array Sequence Analysis/methods , Prenatal Diagnosis/methods , Trisomy/diagnosis , Adult , Aneuploidy , DNA/blood , Female , Humans , Pregnancy , Sequence Analysis, DNA , Trisomy/genetics
6.
Breast Cancer Res ; 14(1): R27, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22321971

ABSTRACT

INTRODUCTION: Although a high frequency of androgen receptor (AR) expression in human breast cancers has been described, exploiting this knowledge for therapy has been challenging. This is in part because androgens can either inhibit or stimulate cell proliferation in pre-clinical models of breast cancer. In addition, many breast cancers co-express other steroid hormone receptors that can affect AR signaling, further obfuscating the effects of androgens on breast cancer cells. METHODS: To create better-defined models of AR signaling in human breast epithelial cells, we took estrogen receptor (ER)-α-negative and progesterone receptor (PR)-negative human breast epithelial cell lines, both cancerous and non-cancerous, and engineered them to express AR, thus allowing the unambiguous study of AR signaling. We cloned a full-length cDNA of human AR, and expressed this transgene in MCF-10A non-tumorigenic human breast epithelial cells and MDA-MB-231 human breast-cancer cells. We characterized the responses to AR ligand binding using various assays, and used isogenic MCF-10A p21 knock-out cell lines expressing AR to demonstrate the requirement for p21 in mediating the proliferative responses to AR signaling in human breast epithelial cells. RESULTS: We found that hyperactivation of the mitogen-activated protein kinase (MAPK) pathway from both AR and epidermal growth factor receptor (EGFR) signaling resulted in a growth-inhibitory response, whereas MAPK signaling from either AR or EGFR activation resulted in cellular proliferation. Additionally, p21 gene knock-out studies confirmed that AR signaling/activation of the MAPK pathway is dependent on p21. CONCLUSIONS: These studies present a new model for the analysis of AR signaling in human breast epithelial cells lacking ERα/PR expression, providing an experimental system without the potential confounding effects of ERα/PR crosstalk. Using this system, we provide a mechanistic explanation for previous observations ascribing a dual role for AR signaling in human breast cancer cells. As previous reports have shown that approximately 40% of breast cancers can lack p21 expression, our data also identify potential new caveats for exploiting AR as a target for breast cancer therapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , MAP Kinase Signaling System , Receptors, Androgen/physiology , Androgen Antagonists/pharmacology , Androgens/pharmacology , Anilides/pharmacology , Breast Neoplasms , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/physiology , Estrogen Receptor alpha/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression , Humans , Metribolone/pharmacology , Nitriles/pharmacology , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Tosyl Compounds/pharmacology , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 106(8): 2835-40, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19196980

ABSTRACT

The phosphatidylinositol 3-kinase subunit PIK3CA is frequently mutated in human cancers. Here we used gene targeting to "knock in" PIK3CA mutations into human breast epithelial cells to identify new therapeutic targets associated with oncogenic PIK3CA. Mutant PIK3CA knockin cells were capable of epidermal growth factor and mTOR-independent cell proliferation that was associated with AKT, ERK, and GSK3beta phosphorylation. Paradoxically, the GSK3beta inhibitors lithium chloride and SB216763 selectively decreased the proliferation of human breast and colorectal cancer cell lines with oncogenic PIK3CA mutations and led to a decrease in the GSK3beta target gene CYCLIN D1. Oral treatment with lithium preferentially inhibited the growth of nude mouse xenografts of HCT-116 colon cancer cells with mutant PIK3CA compared with isogenic HCT-116 knockout cells containing only wild-type PIK3CA. Our findings suggest GSK3beta is an important effector of mutant PIK3CA, and that lithium, an FDA-approved therapy for bipolar disorders, has selective antineoplastic properties against cancers that harbor these mutations.


Subject(s)
Mutation , Oncogenes , Phosphatidylinositol 3-Kinases/genetics , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knock-In Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mammary Glands, Human/metabolism , Mice , Mice, Nude , Phosphorylation , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases , Transplantation, Heterologous
8.
Oncotarget ; 7(5): 6281-93, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26823390

ABSTRACT

Ki-67 expression is correlated with cell proliferation and is a prognostic marker for various cancers; however, its function is unknown. Here we demonstrate that genetic disruption of Ki-67 in human epithelial breast and colon cancer cells depletes the cancer stem cell niche. Ki-67 null cells had a proliferative disadvantage compared to wildtype controls in colony formation assays and displayed increased sensitivity to various chemotherapies. Ki-67 null cancer cells showed decreased and delayed tumor formation in xenograft assays, which was associated with a reduction in cancer stem cell markers. Immunohistochemical analyses of human breast cancers revealed that Ki-67 expression is maintained at equivalent or greater levels in metastatic sites of disease compared to matched primary tumors, suggesting that maintenance of Ki-67 expression is associated with metastatic/clonogenic potential. These results elucidate Ki-67's role in maintaining the cancer stem cell niche, which has potential diagnostic and therapeutic implications for human malignancies.


Subject(s)
Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , Ki-67 Antigen/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction
10.
Nat Commun ; 5: 4961, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25247763

ABSTRACT

The PIK3CA gene is frequently mutated in human cancers. Here we carry out a SILAC-based quantitative phosphoproteomic analysis using isogenic knockin cell lines containing 'driver' oncogenic mutations of PIK3CA to dissect the signalling mechanisms responsible for oncogenic phenotypes induced by mutant PIK3CA. From 8,075 unique phosphopeptides identified, we observe that aberrant activation of PI3K pathway leads to increased phosphorylation of a surprisingly wide variety of kinases and downstream signalling networks. Here, by integrating phosphoproteomic data with human protein microarray-based AKT1 kinase assays, we discover and validate six novel AKT1 substrates, including cortactin. Through mutagenesis studies, we demonstrate that phosphorylation of cortactin by AKT1 is important for mutant PI3K-enhanced cell migration and invasion. Our study describes a quantitative and global approach for identifying mutation-specific signalling events and for discovering novel signalling molecules as readouts of pathway activation or potential therapeutic targets.


Subject(s)
Cortactin/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoproteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Chromatography, Liquid , Class I Phosphatidylinositol 3-Kinases , DNA Primers/genetics , Fluorescent Antibody Technique , Gene Knock-In Techniques , Humans , Immunoblotting , Immunoprecipitation , Mutagenesis, Site-Directed , Mutation/genetics , Proteomics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Tandem Mass Spectrometry
11.
Cancer Res ; 73(11): 3248-61, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23580570

ABSTRACT

The selective pressures leading to cancers with mutations in both KRAS and PIK3CA are unclear. Here, we show that somatic cell knockin of both KRAS G12V and oncogenic PIK3CA mutations in human breast epithelial cells results in cooperative activation of the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways in vitro, and leads to tumor formation in immunocompromised mice. Xenografts from double-knockin cells retain single copies of mutant KRAS and PIK3CA, suggesting that tumor formation does not require increased copy number of either oncogene, and these results were also observed in human colorectal cancer specimens. Mechanistically, the cooperativity between mutant KRAS and PIK3CA is mediated in part by Ras/p110α binding, as inactivating point mutations within the Ras-binding domain of PIK3CA significantly abates pathway signaling. In addition, Pdk1 activation of the downstream effector p90RSK is also increased by the combined presence of mutant KRAS and PIK3CA. These results provide new insights into mutant KRAS function and its role in carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Epithelial Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/pathology , Class I Phosphatidylinositol 3-Kinases , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Female , Gene Knock-In Techniques , Heterografts , Humans , Immunocompromised Host , MAP Kinase Signaling System , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Point Mutation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , ras Proteins/metabolism
12.
Cancer Biol Ther ; 11(3): 358-67, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21124076

ABSTRACT

A high frequency of somatic mutations has been found in breast cancers within the gene encoding the catalytic p110α subunit of PI3K, PIK3CA. Using isogenic human breast epithelial cells, we have previously demonstrated that oncogenic PIK3CA "hotspot" mutations predict for response to the toxic effects of lithium. However, other somatic genetic alterations occur within this pathway in breast cancers, and it is possible that these changes may also predict for lithium sensitivity. We overexpressed the epidermal growth factor receptor (EGFR) into the non-tumorigenic human breast epithelial cell line MCF-10A, and compared these cells to isogenic cell lines previously created via somatic cell gene targeting to model Pten loss, PIK3CA mutations, and the invariant AKT1 mutation, E17K. EGFR overexpressing clones were capable of cellular proliferation in the absence of EGF and were sensitive to lithium similar to the results previously seen with cells harboring PIK3CA mutations. In contrast, AKT1 E17K cells and PTEN -/- cells displayed resistance or partial sensitivity to lithium, respectively. Western blot analysis demonstrated that lithium sensitivity correlated with significant decreases in both PI3K and MAPK signaling that were observed only in EGFR overexpressing and mutant PIK3CA cell lines. These studies demonstrate that EGFR overexpression and PIK3CA mutations are predictors of response to lithium, whereas Pten loss and AKT1 E17K mutations do not predict for lithium sensitivity. Our findings may have important implications for the use of these genetic lesions in breast cancer patients as predictive markers of response to emerging PI3K pathway inhibitors.


Subject(s)
Breast/drug effects , Epithelial Cells/drug effects , ErbB Receptors/genetics , Lithium Compounds/pharmacology , Lithium Compounds/therapeutic use , Phosphatidylinositol 3-Kinases/genetics , Breast/metabolism , Cell Line , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Female , Gene Expression , Humans , Immunoblotting , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
13.
J Clin Invest ; 120(8): 2655-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20664174

ABSTRACT

Targeted cancer therapeutics can be effective when patients are preselected to maximize the chance of response. Increasingly, molecular markers such as oncogenic DNA mutations are being exploited to help guide patient preselection. These DNA lesions can predict for either a positive or negative response to a given drug. Finding such predictive biomarkers is an ongoing challenge. New work by Di Nicolantonio and colleagues in this issue of the JCI demonstrates that PI3K catalytic alpha subunit (PIK3CA) mutations can sensitize cancer cells to the mammalian target of rapamycin (mTOR) inhibitor everolimus. In addition, they show that the concurrent presence of PIK3CA mutations and mutations in either KRAS or BRAF predict for resistance to this drug. These data suggest that mTOR inhibitors currently in use will be ineffective against cancers that have a mutation in either KRAS or BRAF despite having PI3K/AKT/mTOR pathway activation.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mutation , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Sirolimus/analogs & derivatives , ras Proteins/genetics , Class I Phosphatidylinositol 3-Kinases , Everolimus , Humans , Intracellular Signaling Peptides and Proteins/physiology , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL