Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
2.
IEEE Trans Cybern ; 51(12): 5671-5680, 2021 Dec.
Article in English | MEDLINE | ID: mdl-31794411

ABSTRACT

Effectively recognizing different sceneries with complex backgrounds and varied lighting conditions plays an important role in modern AI systems. Competitive performance has recently been achieved by the deep scene categorization models. However, these models implicitly hypothesize that the image-level labels are 100% correct, which is too restrictive. Practically, the image-level labels for massive-scale scenery sets are usually calculated by external predictors such as ImageNet-CN. These labels can easily become contaminated because no predictors are completely accurate. This article proposes a new deep architecture that calculates scene categories by hierarchically deriving stable templates, which are discovered using a generative model. Specifically, we first construct a semantic space by incorporating image-level labels using subspace embedding. Afterward, it is noticeable that in the semantic space, the superpixel distributions from identically labeled images remain unchanged, regardless of the image-level label noises. On the basis of this observation, a probabilistic generative model learns the stable templates for each scene category. To deeply represent each scenery category, a novel aggregation network is developed to statistically concatenate the CNN features learned from scene annotations predicted by HSA. Finally, the learned deep representations are integrated into an image kernel, which is subsequently incorporated into a multiclass SVM for distinguishing scene categories. Thorough experiments have shown the performance of our method. As a byproduct, an empirical study of 33 SIFT-flow categories shows that the learned stable templates remain almost unchanged under a nearly 36% image label contamination rate.


Subject(s)
Semantics
3.
Oncogene ; 38(22): 4232-4249, 2019 05.
Article in English | MEDLINE | ID: mdl-30718920

ABSTRACT

Lysine methylation of histones and non-histone substrates by the SET domain containing protein lysine methyltransferase (KMT) G9a/EHMT2 governs transcription contributing to apoptosis, aberrant cell growth, and pluripotency. The positioning of chromosomes within the nuclear three-dimensional space involves interactions between nuclear lamina (NL) and the lamina-associated domains (LAD). Contact of individual LADs with the NL are dependent upon H3K9me2 introduced by G9a. The mechanisms governing the recruitment of G9a to distinct subcellular sites, into chromatin or to LAD, is not known. The cyclin D1 gene product encodes the regulatory subunit of the holoenzyme that phosphorylates pRB and NRF1 thereby governing cell-cycle progression and mitochondrial metabolism. Herein, we show that cyclin D1 enhanced H3K9 dimethylation though direct association with G9a. Endogenous cyclin D1 was required for the recruitment of G9a to target genes in chromatin, for G9a-induced H3K9me2 of histones, and for NL-LAD interaction. The finding that cyclin D1 is required for recruitment of G9a to target genes in chromatin and for H3K9 dimethylation, identifies a novel mechanism coordinating protein methylation.


Subject(s)
Cyclin D1/metabolism , DNA Methylation/physiology , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Chromosomes/physiology , HEK293 Cells , Humans , MCF-7 Cells , Protein Binding/physiology
4.
Theranostics ; 8(8): 2251-2263, 2018.
Article in English | MEDLINE | ID: mdl-29721077

ABSTRACT

Background: Genetic classification of breast cancer based on the coding mRNA suggests the evolution of distinct subtypes. Whether the non-coding genome is altered concordantly with the coding genome and the mechanism by which the cell cycle directly controls the non-coding genome is poorly understood. Methods: Herein, the miRNA signature maintained by endogenous cyclin D1 in human breast cancer cells was defined. In order to determine the clinical significance of the cyclin D1-mediated miRNA signature, we defined a miRNA expression superset from 459 breast cancer samples. We compared the coding and non-coding genome of breast cancer subtypes. Results: Hierarchical clustering of human breast cancers defined four distinct miRNA clusters (G1-G4) associated with distinguishable relapse-free survival by Kaplan-Meier analysis. The cyclin D1-regulated miRNA signature included several oncomirs, was conserved in multiple breast cancer cell lines, was associated with the G2 tumor miRNA cluster, ERα+ status, better outcome and activation of the Wnt pathway. The coding and non-coding genome were discordant within breast cancer subtypes. Seed elements for cyclin D1-regulated miRNA were identified in 63 genes of the Wnt signaling pathway including DKK. Cyclin D1 restrained DKK1 via the 3'UTR. In vivo studies using inducible transgenics confirmed cyclin D1 induces Wnt-dependent gene expression. Conclusion: The non-coding genome defines breast cancer subtypes that are discordant with their coding genome subtype suggesting distinct evolutionary drivers within the tumors. Cyclin D1 orchestrates expression of a miRNA signature that induces Wnt/ß-catenin signaling, therefore cyclin D1 serves both upstream and downstream of Wnt/ß-catenin signaling.


Subject(s)
Breast Neoplasms/genetics , Cyclin D1/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Animals , Cyclin D1/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Mice , MicroRNAs/metabolism , Prognosis , Treatment Outcome , Wnt Signaling Pathway/genetics
5.
Oncotarget ; 8(47): 81754-81775, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29137220

ABSTRACT

The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that drives cell autonomous cell cycle progression and proliferation. Herein we show cyclin D1 abundance is increased >30-fold in the stromal fibroblasts of patients with invasive breast cancer, associated with poor outcome. Cyclin D1 transformed hTERT human fibroblast to a cancer-associated fibroblast phenotype. Stromal fibroblast expression of cyclin D1 (cyclin D1Stroma) in vivo, enhanced breast epithelial cancer tumor growth, restrained apoptosis, and increased autophagy. Cyclin D1Stroma had profound effects on the breast tumor microenvironment increasing the recruitment of F4/80+ and CD11b+ macrophages and increasing angiogenesis. Cyclin D1Stroma induced secretion of factors that promoted expansion of stem cells (breast stem-like cells, embryonic stem cells and bone marrow derived stem cells). Cyclin D1Stroma resulted in increased secretion of proinflammatory cytokines (CCL2, CCL7, CCL11, CXCL1, CXCL5, CXCL9, CXCL12), CSF (CSF1, GM-CSF1) and osteopontin (OPN) (30-fold). OPN was induced by cyclin D1 in fibroblasts, breast epithelial cells and in the murine transgenic mammary gland and OPN was sufficient to induce stem cell expansion. These results demonstrate that cyclin D1Stroma drives tumor microenvironment heterocellular signaling, promoting several key hallmarks of cancer.

6.
Cancer Res ; 76(22): 6723-6734, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27634768

ABSTRACT

Proteomic analysis of castration-resistant prostate cancer demonstrated the enrichment of Src tyrosine kinase activity in approximately 90% of patients. Src is known to induce cyclin D1, and a cyclin D1-regulated gene expression module predicts poor outcome in human prostate cancer. The tumor-associated calcium signal transducer 2 (TACSTD2/Trop2/M1S1) is enriched in the prostate, promoting prostate stem cell self-renewal upon proteolytic activation via a γ-secretase cleavage complex (PS1, PS2) and TACE (ADAM17), which releases the Trop2 intracellular domain (Trop2 ICD). Herein, v-Src transformation of primary murine prostate epithelial cells increased the proportion of prostate cancer stem cells as characterized by gene expression, epitope characteristics, and prostatosphere formation. Cyclin D1 was induced by v-Src, and Src kinase induction of Trop2 ICD nuclear accumulation required cyclin D1. Cyclin D1 induced abundance of the Trop2 proteolytic cleavage activation components (PS2, TACE) and restrained expression of the inhibitory component of the Trop2 proteolytic complex (Numb). Patients with prostate cancer with increased nuclear Trop2 ICD and cyclin D1, and reduced Numb, had reduced recurrence-free survival probability (HR = 4.35). Cyclin D1, therefore, serves as a transducer of v-Src-mediated induction of Trop2 ICD by enhancing abundance of the Trop2 proteolytic activation complex. Cancer Res; 76(22); 6723-34. ©2016 AACR.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cyclin D1/metabolism , src-Family Kinases/metabolism , Animals , Humans , Mice , Signal Transduction , Transfection
8.
Oncotarget ; 7(5): 5383-400, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26689991

ABSTRACT

Patients with hormone-resistant prostate cancer (PCa) have higher biochemical failure rates following radiation therapy (RT). Cyclin D1 deregulated expression in PCa is associated with a more aggressive disease: however its role in radioresistance has not been determined. Cyclin D1 levels in the androgen-independent PC3 and 22Rv1 PCa cells were stably inhibited by infecting with cyclin D1-shRNA. Tumorigenicity and radiosensitivity were investigated using in vitro and in vivo experimental assays. Cyclin D1 silencing interfered with PCa oncogenic phenotype by inducing growth arrest in the G1 phase of cell cycle and reducing soft agar colony formation, migration, invasion in vitro and tumor formation and neo-angiogenesis in vivo. Depletion of cyclin D1 significantly radiosensitizes PCa cells by increasing the RT-induced DNA damages by affecting the NHEJ and HR pathways responsible of the DNA double-strand break repair. Following treatment of cells with RT the abundance of a biomarker of DNA damage, γ-H2AX, was dramatically increased in sh-cyclin D1 treated cells compared to shRNA control. Concordant with these observations DNA-PKcs-activation and RAD51-accumulation, part of the DNA double-strand break repair machinery, were reduced in shRNA-cyclin D1 treated cells compared to shRNA control. We further demonstrate the physical interaction between CCND1 with activated-ATM, -DNA-PKcs and RAD51 is enhanced by RT. Finally, siRNA-mediated silencing experiments indicated DNA-PKcs and RAD51 are downstream targets of CCND1-mediated PCa cells radioresistance. In summary, these observations suggest that CCND1 is a key mediator of PCa radioresistance and could represent a potential target for radioresistant hormone-resistant PCa.


Subject(s)
Cyclin D1/antagonists & inhibitors , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , Prostatic Neoplasms, Castration-Resistant/prevention & control , RNA, Small Interfering/genetics , Radiation Tolerance/genetics , Radiation-Sensitizing Agents , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , DNA Repair/radiation effects , Fluorescent Antibody Technique , Histones/metabolism , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Phosphorylation/radiation effects , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction/radiation effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cancer Res ; 76(2): 329-38, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26582866

ABSTRACT

Therapy resistance and poor outcome in prostate cancer is associated with increased expression of cyclin D1. Androgens promote DNA double-strand break repair to reduce DNA damage, and cyclin D1 was also shown to enhance DNA damage repair (DDR). In this study, we investigated the significance of cyclin D1 in androgen-induced DDR using established prostate cancer cells and prostate tissues from cyclin D1 knockout mice. We demonstrate that endogenous cyclin D1 further diminished the dihydrotestosterone (DHT)-dependent reduction of γH2AX foci in vitro. We also show that cyclin D1 was required for the androgen-dependent DNA damage response both in vitro and in vivo. Furthermore, cyclin D1 was required for androgen-enhanced DDR and radioresistance of prostate cancer cells. Moreover, microarray analysis of primary prostate epithelial cells from cyclin D1-deficient and wild-type mice demonstrated that most of the DHT-dependent gene expression changes are also cyclin D1 dependent. Collectively, our findings suggest that the hormone-mediated recruitment of cyclin D1 to sites of DDR may facilitate the resistance of prostate cancer cells to DNA damage therapies and highlight the need to explore other therapeutic approaches in prostate cancer to prevent or overcome drug resistance.


Subject(s)
Cyclin D1/genetics , DNA Damage , DNA Repair , Dihydrotestosterone/pharmacology , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Animals , Cell Line, Tumor , Cyclin D1/biosynthesis , Histones/genetics , Histones/metabolism , Humans , Male , Mice , Mice, Knockout , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Transfection
10.
Cancer Res ; 75(10): 1992-2004, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25769723

ABSTRACT

Prostate cancer is the second leading form of cancer-related death in men. In a subset of prostate cancer patients, increased chemokine signaling IL8 and IL6 correlates with castrate-resistant prostate cancer (CRPC). IL8 and IL6 are produced by prostate epithelial cells and promote prostate cancer cell invasion; however, the mechanisms restraining prostate epithelial cell cytokine secretion are poorly understood. Herein, the cell-fate determinant factor DACH1 inhibited CRPC tumor growth in mice. Using Dach1(fl/fl)/Probasin-Cre bitransgenic mice, we show IL8 and IL6 secretion was altered by approximately 1,000-fold by endogenous Dach1. Endogenous Dach1 is shown to serve as a key endogenous restraint to prostate epithelial cell growth and restrains migration via CXCL signaling. DACH1 inhibited expression, transcription, and secretion of the CXCL genes (IL8 and IL6) by binding to their promoter regulatory regions in chromatin. DACH1 is thus a newly defined determinant of benign and malignant prostate epithelium cellular growth, migration, and cytokine abundance in vivo.


Subject(s)
Cell Movement , Cytokines/metabolism , Epithelial Cells/physiology , Eye Proteins/physiology , Prostatic Neoplasms/metabolism , Transcription Factors/physiology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Male , Mice, Transgenic , Neoplasm Transplantation , Prostate/pathology , Prostatic Neoplasms/pathology , Protein Sorting Signals
11.
Cancer Res ; 74(23): 7103-14, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25452256

ABSTRACT

Src family kinases (SFK) integrate signal transduction for multiple receptors, regulating cellular proliferation, invasion, and metastasis in human cancer. Although Src is rarely mutated in human prostate cancer, SFK activity is increased in the majority of human prostate cancers. To determine the molecular mechanisms governing prostate cancer bone metastasis, FVB murine prostate epithelium was transduced with oncogenic v-Src. The prostate cancer cell lines metastasized in FVB mice to brain and bone. Gene expression profiling of the tumors identified activation of a CCR5 signaling module when the prostate epithelial cell lines were grown in vivo versus tissue cultures. The whole body, bone, and brain metastatic prostate cancer burden was reduced by oral CCR5 antagonist. Clinical trials of CCR5 inhibitors may warrant consideration in patients with CCR5 activation in their tumors.


Subject(s)
Bone Neoplasms/prevention & control , CCR5 Receptor Antagonists/pharmacology , Genes, src , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Receptors, CCR5/genetics , src-Family Kinases/genetics , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Brain Neoplasms/metabolism , Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Cell Line, Transformed , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Gene Expression Profiling/methods , Humans , Male , Mice , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptors, CCR5/metabolism , src-Family Kinases/metabolism
12.
Cancer Res ; 74(14): 3959-70, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24830723

ABSTRACT

The cyclin D1 gene encodes the regulatory subunit of a holoenyzme that phosphorylates the retinoblastoma protein (pRb) and nuclear respiratory factor (NRF1) proteins. The abundance of cyclin D1 determines estrogen-dependent gene expression in the mammary gland of mice. Using estradiol (E2) and an E2-dendrimer conjugate that is excluded from the nucleus, we demonstrate that E2 delays the DNA damage response (DDR) via an extranuclear mechanism. The E2-induced DDR required extranuclear cyclin D1, which bound ERα at the cytoplasmic membrane and augmented AKT phosphorylation (Ser473) and γH2AX foci formation. In the nucleus, E2 inhibited, whereas cyclin D1 enhanced homology-directed DNA repair. Cyclin D1 was recruited to γH2AX foci by E2 and induced Rad51 expression. Cyclin D1 governs an essential role in the E2-dependent DNA damage response via a novel extranuclear function. The dissociable cytoplasmic function to delay the E2-mediated DDR together with the nuclear enhancement of DNA repair uncovers a novel extranuclear function of cyclin D1 that may contribute to the role of E2 in breast tumorigenesis.


Subject(s)
Cyclin D1/genetics , DNA Repair , Estrogens/metabolism , Signal Transduction , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatin/metabolism , Cyclin D1/metabolism , DNA Damage , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Mice , Protein Binding , RNA Interference , Rad51 Recombinase/metabolism
13.
Oncotarget ; 5(4): 1083-90, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24658544

ABSTRACT

The serine threonine kinase Akt1 has been implicated in the control of cellular metabolism, survival and growth. Herein, disruption of the ubiquitously expressed member of the Akt family of genes, Akt1, in the mouse, demonstrates a requirement for Akt1 in miRNA-mediated cellular apoptosis. The miR-17/20 cluster is known to inhibit breast cancer cellular proliferation through G1/S cell cycle arrest via binding to the cyclin D1 3'UTR. Here we show that miR-17/20 overexpression sensitizes cells to apoptosis induced by either Doxorubicin or UV irradiation in MCF-7 cells via Akt1. miR-17/20 mediates apoptosis via increased p53 expression which promotes Akt degradation. Akt1⁻/⁻ mammary epithelial cells which express Akt2 and Akt3 demonstrated increased apoptosis to DNA damaging agents. Akt1 deficiency abolished the miR-17/20-mediated apoptosis. These results demonstrated a novel pathway through which miR17/20 regulate p53 and Akt controlling breast cancer cell apoptosis.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Doxorubicin/pharmacology , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Prognosis
14.
Cancer Res ; 74(2): 508-19, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24282282

ABSTRACT

Improved clinical management of prostate cancer has been impeded by an inadequate understanding of molecular genetic elements governing tumor progression. Gene signatures have provided improved prognostic indicators of human prostate cancer. The TGF-ß/BMP-SMAD4 signaling pathway, which induces epithelial-mesenchymal transition (EMT), is known to constrain prostate cancer progression induced by Pten deletion. Herein, cyclin D1 inactivation reduced cellular proliferation in the murine prostate in vivo and in isogenic oncogene-transformed prostate cancer cell lines. The in vivo cyclin D1-mediated molecular signature predicted poor outcome of recurrence-free survival for patients with prostate cancer (K-means HR, 3.75, P = 0.02) and demonstrated that endogenous cyclin D1 restrains TGF-ß, Snail, Twist, and Goosecoid signaling. Endogenous cyclin D1 enhanced Wnt and ES cell gene expression and expanded a prostate stem cell population. In chromatin immunoprecipitation sequencing, cyclin D1 occupied genes governing stem cell expansion and induced their transcription. The coordination of EMT restraining and stem cell expanding gene expression by cyclin D1 in the prostate may contribute to its strong prognostic value for poor outcome in biochemical-free recurrence in human prostate cancer.


Subject(s)
Cyclin D1/physiology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Disease-Free Survival , Gene Deletion , Humans , Male , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/metabolism , Prognosis , Recurrence , Signal Transduction , Treatment Outcome
15.
Nat Commun ; 4: 2812, 2013.
Article in English | MEDLINE | ID: mdl-24287487

ABSTRACT

Cyclin D1 encodes the regulatory subunit of a holoenzyme that phosphorylates the pRB protein and promotes G1/S cell-cycle progression and oncogenesis. Dicer is a central regulator of miRNA maturation, encoding an enzyme that cleaves double-stranded RNA or stem-loop-stem RNA into 20-25 nucleotide long small RNA, governing sequence-specific gene silencing and heterochromatin methylation. The mechanism by which the cell cycle directly controls the non-coding genome is poorly understood. Here we show that cyclin D1(-/-) cells are defective in pre-miRNA processing which is restored by cyclin D1a rescue. Cyclin D1 induces Dicer expression in vitro and in vivo. Dicer is transcriptionally targeted by cyclin D1, via a cdk-independent mechanism. Cyclin D1 and Dicer expression significantly correlates in luminal A and basal-like subtypes of human breast cancer. Cyclin D1 and Dicer maintain heterochromatic histone modification (Tri-m-H3K9). Cyclin D1-mediated cellular proliferation and migration is Dicer-dependent. We conclude that cyclin D1 induction of Dicer coordinates microRNA biogenesis.


Subject(s)
Breast Neoplasms/metabolism , Cyclin D1/physiology , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/metabolism , MicroRNAs/biosynthesis , Ribonuclease III/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Movement/genetics , Cell Proliferation , Female , HCT116 Cells , Histones/metabolism , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Protein Processing, Post-Translational/genetics
16.
Sci Rep ; 3: 1697, 2013.
Article in English | MEDLINE | ID: mdl-23603888

ABSTRACT

Using a newly developed near-infrared (NIR) dye that fluoresces at two different wavelengths (dichromic fluorescence, DCF), we discovered a new fluorescent substrate for Akt, also known as protein kinase B, and a method to quantitatively report this enzyme's activity in real time. Upon insulin activation of cellular Akt, the enzyme multi-phosphorylated a single serine residue of a diserine DCF substrate in a time-dependent manner, culminating in monophospho- to triphospho-serine products. The NIR DCF probe was highly selective for the Akt1 isoform, which was demonstrated using Akt1 knockout cells derived from MMTV-ErbB2 transgenic mice. The DCF mechanism provides unparalleled potential to assess the stimulation, sustainability, and reversibility of Akt activation longitudinally. Importantly, NIR fluorescence provides a pathway to translate findings from cells to living organisms, a condition that could eventually facilitate the use of these probes in humans.


Subject(s)
Fluorescent Dyes , Gene Expression Profiling/methods , Microscopy, Fluorescence, Multiphoton/methods , Oncogene Protein v-akt/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Enzyme Activation , MCF-7 Cells , Mice
17.
Cancer Res ; 73(2): 978-89, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23204233

ABSTRACT

Herein, murine prostate cancer cell lines, generated via selective transduction with a single oncogene (c-Myc, Ha-Ras, and v-Src), showed oncogene-specific prostate cancer molecular signatures that were recapitulated in human prostate cancer and developed lung metastasis in immune-competent mice. Interrogation of two independent retrospective cohorts of patient samples using the oncogene signature showed an ability to distinguish tumor from normal prostate with a predictive value for prostate cancer of 98% to 99%. In a blinded study, the signature algorithm showed independent substratification of reduced recurrence-free survival by Kaplan-Meier analysis. The generation of new oncogene-specific prostate cancer cell lines that recapitulate human prostate cancer gene expression, which metastasize in immune-competent mice, are a valuable new resource for testing targeted therapy, whereas the molecular signatures identified herein provides further value over current gene signature markers of prediction and outcome.


Subject(s)
Cell Line, Tumor , Neoplasm Metastasis , Oncogenes , Prostatic Neoplasms/genetics , Animals , Cell Transformation, Neoplastic , Disease Progression , Disease-Free Survival , Genes, myc , Genes, ras , Genes, src , Lung Neoplasms/secondary , Male , Mice , Mice, Transgenic , Predictive Value of Tests , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Transcriptome
18.
Cancer Res ; 70(24): 10464-73, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159656

ABSTRACT

The role of mammary epithelial cell (MEC) NF-κB in tumor progression in vivo is unknown, as murine NF-κB components and kinases either are required for murine survival or interfere with normal mammary gland development. As NF-κB inhibitors block both tumor-associated macrophages (TAM) and MEC NF-κB, the importance of MEC NF-κB to tumor progression in vivo remained to be determined. Herein, an MEC-targeted inducible transgenic inhibitor of NF-κB (IκBαSR) was developed in ErbB2 mammary oncomice. Inducible suppression of NF-κB in the adult mammary epithelium delayed the onset and number of new tumors. Within similar sized breast tumors, TAM and tumor neoangiogenesis was reduced. Coculture experiments demonstrated MEC NF-κB enhanced TAM recruitment. Genome-wide expression and proteomic analysis showed that IκBαSR inhibited tumor stem cell pathways. IκBαSR inhibited breast tumor stem cell markers in transgenic tumors, reduced stem cell expansion in vitro, and repressed expression of Nanog and Sox2 in vivo and in vitro. MEC NF-κB contributes to mammary tumorigenesis. As we show that NF-κB contributes to expansion of breast tumor stem cells and heterotypic signals that enhance TAM and vasculogenesis, these processes may contribute to NF-κB-dependent mammary tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/pathology , Mammary Neoplasms, Experimental/pathology , NF-kappa B/metabolism , Neoplastic Stem Cells/pathology , Animals , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Female , I-kappa B Proteins/biosynthesis , I-kappa B Proteins/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Neoplastic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/biosynthesis , Transfection
19.
Mol Biol Cell ; 21(23): 4264-74, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926681

ABSTRACT

c-jun, which is overexpressed in a number of human cancers encodes a critical component of the AP-1 complex. c-jun has been shown to either induce or inhibit cellular apoptosis. Germ line deletion of both c-jun alleles is embryonically lethal. To determine the role of the endogenous c-jun gene in apoptosis, we performed mammary epithelial cell-targeted somatic deletion using floxed c-jun (c-jun(f/f)) conditional knockout mice. Laser capture microdissection demonstrated endogenous c-jun inhibits expression of apoptosis inducing genes and reactive oxygen species (ROS)-reducing genes (MnSOD, catalase). ROS have been implicated in apoptosis and undergo enzymatic elimination via MnSOD and CuZnSOD with further detoxification via catalase. c-jun-mediated survival was in part dependent on ROS production. c-jun-mediated repression of MnSOD and catalase occurred via mitochondrial complex I and NOX I. Collectively, these studies define a pivotal role of endogenous c-jun in promoting cell survival via maintaining mitochondrial integrity and expression of the key regulators of ROS production.


Subject(s)
Apoptosis , Genes, jun , Mammary Glands, Animal/cytology , NADH, NADPH Oxidoreductases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Reactive Oxygen Species/metabolism , Animals , Catalase/metabolism , Cell Survival , Electron Transport Complex I/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Gene Expression Regulation , Glutathione Peroxidase/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Mammary Glands, Animal/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Mice, Transgenic , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Repressor Proteins/metabolism , Signal Transduction/genetics , Superoxide Dismutase/metabolism , Survivin , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...