Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Proc Natl Acad Sci U S A ; 116(15): 7353-7362, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30910979

ABSTRACT

Carcinoma cells residing in an intermediate phenotypic state along the epithelial-mesenchymal (E-M) spectrum are associated with malignant phenotypes, such as invasiveness, tumor-initiating ability, and metastatic dissemination. Using the recently described CD104+/CD44hi antigen marker combination, we isolated highly tumorigenic breast cancer cells residing stably-both in vitro and in vivo-in an intermediate phenotypic state and coexpressing both epithelial (E) and mesenchymal (M) markers. We demonstrate that tumorigenicity depends on individual cells residing in this E/M hybrid state and cannot be phenocopied by mixing two cell populations that reside stably at the two ends of the spectrum, i.e., in the E and in the M state. Hence, residence in a specific intermediate state along the E-M spectrum rather than phenotypic plasticity appears critical to the expression of tumor-initiating capacity. Acquisition of this E/M hybrid state is facilitated by the differential expression of EMT-inducing transcription factors (EMT-TFs) and is accompanied by the expression of adult stem cell programs, notably, active canonical Wnt signaling. Furthermore, transition from the highly tumorigenic E/M state to a fully mesenchymal phenotype, achieved by constitutive ectopic expression of Zeb1, is sufficient to drive cells out of the E/M hybrid state into a highly mesenchymal state, which is accompanied by a substantial loss of tumorigenicity and a switch from canonical to noncanonical Wnt signaling. Identifying the gatekeepers of the various phenotypic states arrayed along the E-M spectrum is likely to prove useful in developing therapeutic approaches that operate by shifting cancer cells between distinct states along this spectrum.


Subject(s)
Adult Stem Cells/metabolism , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplasms, Basal Cell/metabolism , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway , Adult Stem Cells/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/pathology , Neoplastic Stem Cells/pathology
2.
Chemistry ; 27(1): 362-370, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-32716591

ABSTRACT

During the last decades, photodynamic therapy (PDT), an approved medical technique, has received increasing attention to treat certain types of cancer. Despite recent improvements, the treatment of large tumors remains a major clinical challenge due to the low ability of the photosensitizer (PS) to penetrate a 3D cellular architecture and the low oxygen concentrations present in the tumor center. To mimic the conditions found in clinical tumors, exceptionally large 3D multicellular tumor spheroids (MCTSs) with a diameter of 800 µm were used in this work to test a series of new RuII polypyridine complexes as one-photon and two-photon PSs. These metal complexes were found to fully penetrate the 3D cellular architecture and to generate singlet oxygen in the hypoxic center upon light irradiation. While having no observed dark toxicity, the lead compound of this study showed an impressive phototoxicity upon clinically relevant one-photon (595 nm) or two-photon (800 nm) excitation with a full eradication of the hypoxic center of the MCTSs. Importantly, this efficacy was also demonstrated on mice bearing an adenocarcinomic human alveolar basal epithelial tumor.


Subject(s)
Organometallic Compounds , Photochemotherapy , Photosensitizing Agents , Ruthenium , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Animals , HeLa Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Neoplasms, Basal Cell/drug therapy , Neoplasms, Basal Cell/metabolism , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Photons/therapeutic use , Photosensitizing Agents/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology , Singlet Oxygen/metabolism , Spheroids, Cellular , Tumor Hypoxia , Xenograft Model Antitumor Assays
3.
Int J Mol Sci ; 22(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299265

ABSTRACT

Smoking is a major risk factor for chronic obstructive pulmonary disease (COPD) and causes remodeling of the small airways. However, the exact smoke-induced effects on the different types of small airway epithelial cells (SAECs) are poorly understood. Here, using air-liquid interface (ALI) cultures, single-cell RNA-sequencing reveals previously unrecognized transcriptional heterogeneity within the small airway epithelium and cell type-specific effects upon acute and chronic cigarette smoke exposure. Smoke triggers detoxification and inflammatory responses and aberrantly activates and alters basal cell differentiation. This results in an increase of inflammatory basal-to-secretory cell intermediates and, particularly after chronic smoke exposure, a massive expansion of a rare inflammatory and squamous metaplasia associated KRT6A+ basal cell state and an altered secretory cell landscape. ALI cultures originating from healthy non-smokers and COPD smokers show similar responses to cigarette smoke exposure, although an increased pro-inflammatory profile is conserved in the latter. Taken together, the in vitro models provide high-resolution insights into the smoke-induced remodeling of the small airways resembling the pathological processes in COPD airways. The data may also help to better understand other lung diseases including COVID-19, as the data reflect the smoke-dependent variable induction of SARS-CoV-2 entry factors across SAEC populations.


Subject(s)
Airway Remodeling/drug effects , Alveolar Epithelial Cells/drug effects , Cigarette Smoking/adverse effects , Epithelial Cells/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Cell Differentiation/drug effects , Cells, Cultured , Cigarette Smoking/metabolism , Epithelial Cells/drug effects , Humans , Neoplasms, Basal Cell/metabolism , Primary Cell Culture , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Smoke , Smoking/adverse effects , Smoking/metabolism
4.
Mol Carcinog ; 58(5): 699-707, 2019 05.
Article in English | MEDLINE | ID: mdl-30604896

ABSTRACT

GRB7 gene encodes a multi-domain signal transduction molecule and is part of the core of the HER-2 amplicon. GRB7 is commonly co-amplified and overexpressed with HER-2 in human breast cancer. This study addresses the role of GRB7 in HER-2 positive human breast cancers resistant to HER-2 targeted therapy. HCC1954, 21MT1, and JIMT1 are basal like HER-2 positive breast cancer cell lines based on expression profiling. These three cell lines are resistant to trastuzumab and lapatinib treatment. Knockdown of GRB7 protein expression with siRNA transfection as well as lentiviral vector mediated shRNA over-expression decreased the growth of HCC1954, 21MT1, and JIMT1 cells in vitro and the growth of tumor xenografts these cells formed in animal models. When assayed by ki-67 staining and TUNEL assay, the mechanism of reduced tumor xenograft growth appeared to be distinct. Reduced proliferation and increased apoptosis were seen in 21MT1 cells, while reduced proliferation was seen in HCC1954 cells and increased apoptosis in JIMT1 cells. Phospho-proteome profiling found HER-1 tyrosine phosphorylation was reduced with GRB7 knock down in JIMT1 cells. Immuno-blotting and immuno-precipitation experiments found HER-1 phosphorylation was reduced with GRB7 knock down in all three cell lines. HER-1 knock down via siRNA transient transfection as well as blocking HER-1 function with panitumumab decreased proliferation of all three cell lines in vitro. Our study finds that GRB7 has an essential growth promoting function which is mediated in part by HER-1 activation. The potential of HER-1 targeting in therapy resistant HER-2 positive breast cancer merits further study.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , GRB7 Adaptor Protein/metabolism , Neoplasms, Basal Cell/pathology , Receptor, ErbB-2/metabolism , Animals , Apoptosis , Breast Neoplasms/metabolism , Cell Movement , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Basal Cell/metabolism , Phosphorylation , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Breast Cancer Res Treat ; 162(3): 439-450, 2017 04.
Article in English | MEDLINE | ID: mdl-28176176

ABSTRACT

PURPOSE: The PI3K/Akt signaling axis contributes to the dysregulation of many dominant features in breast cancer including cell proliferation, survival, metabolism, motility, and genomic instability. While multiple studies have demonstrated that basal-like or triple-negative breast tumors have uniformly high PI3K/Akt activity, genomic alterations that mediate dysregulation of this pathway in this subset of highly aggressive breast tumors remain to be determined. METHODS: In this study, we present an integrated genomic analysis based on the use of a PI3K gene expression signature as a framework to analyze orthogonal genomic data from human breast tumors, including RNA expression, DNA copy number alterations, and protein expression. In combination with data from a genome-wide RNA-mediated interference screen in human breast cancer cell lines, we identified essential genetic drivers of PI3K/Akt signaling. RESULTS: Our in silico analyses identified SOX4 amplification as a novel modulator of PI3K/Akt signaling in breast cancers and in vitro studies confirmed its role in regulating Akt phosphorylation. CONCLUSIONS: Taken together, these data establish a role for SOX4-mediated PI3K/Akt signaling in breast cancer and suggest that SOX4 may represent a novel therapeutic target and/or biomarker for current PI3K family therapies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Amplification , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , SOXC Transcription Factors/genetics , Signal Transduction , Breast Neoplasms/pathology , Cell Line, Tumor , Computational Biology/methods , DNA Copy Number Variations , Databases, Genetic , Female , Gene Expression Profiling , Humans , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/pathology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , SOXC Transcription Factors/metabolism , Transcriptome
6.
Med Sci Monit ; 23: 695-703, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28170369

ABSTRACT

BACKGROUND The human LMO2 gene was first cloned from an acute T lymphocytic leukemia patient; it is primarily expressed in hematopoietic and vascular endothelial systems, and functions as a pivotal transcriptional regulator during embryonic hematopoiesis and angiogenesis. However, some recent reports indicated that LMO2 is widely expressed in many tissues and tumors, predominantly in cytoplasm, and revealed complicated functions on tumor behaviors in a variety of cancer types. As an adaptor molecule, binding partners and function details of LMO2 in these solid tumors need to be further investigated. MATERIAL AND METHODS In this study, we used yeast two-hybrid method to screen potential LMO2 interacting partners, MBP-pulldown, and co-immunoprecipitation assay to confirm protein-protein interactions, and confocal microscopy to reveal the subcellular localization of relevant proteins and actin cytoskeleton changes in relevant cells. RESULTS We found that ARP3 and profilin1 were 2 binding partners of LMO2, primarily in cytoplasm. LMO2. Functionally, LMO2 mediated the assembly of a complex including ARP3, profilin1, and actin monomer, increased actin monomer binding to profilin1, and promoted lamellipodia/filopodia formation in basal-type breast cancer cells. CONCLUSIONS Our data indicate a novel functional mechanism of LMO2 in facilitating the delivery of actin monomers to the branched microfilament and increasing lamellipodia/filopodia formation in basal-type breast cancer cells, suggesting a cancer-promoting role of LMO2 in a subtype-dependent manner and its potential as a subtype-specific biomarker for clinical treatment of breast cancers.


Subject(s)
Actin-Related Protein 3/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , LIM Domain Proteins/metabolism , Neoplasms, Basal Cell/pathology , Profilins/metabolism , Proto-Oncogene Proteins/metabolism , Pseudopodia/metabolism , Actin-Related Protein 3/genetics , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Female , HEK293 Cells , Humans , LIM Domain Proteins/genetics , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/metabolism , Profilins/genetics , Protein Binding , Proto-Oncogene Proteins/genetics , Pseudopodia/genetics , Pseudopodia/pathology , Transfection , Two-Hybrid System Techniques
7.
PLoS Genet ; 10(7): e1004520, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25079073

ABSTRACT

Mammary gland branching morphogenesis and ductal homeostasis relies on mammary stem cell function for the maintenance of basal and luminal cell compartments. The mechanisms of transcriptional regulation of the basal cell compartment are currently unknown. We explored these mechanisms in the basal cell compartment and identified the Co-factor of LIM domains (CLIM/LDB/NLI) as a transcriptional regulator that maintains these cells. Clims act within the basal cell compartment to promote branching morphogenesis by maintaining the number and proliferative potential of basal mammary epithelial stem cells. Clim2, in a complex with LMO4, supports mammary stem cells by directly targeting the Fgfr2 promoter in basal cells to increase its expression. Strikingly, Clims also coordinate basal-specific transcriptional programs to preserve luminal cell identity. These basal-derived cues inhibit epidermis-like differentiation of the luminal cell compartment and enhance the expression of luminal cell-specific oncogenes ErbB2 and ErbB3. Consistently, basal-expressed Clims promote the initiation and progression of breast cancer in the MMTV-PyMT tumor model, and the Clim-regulated branching morphogenesis gene network is a prognostic indicator of poor breast cancer outcome in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , LIM Domain Proteins/genetics , Neoplasms, Basal Cell/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Transcription Factors/genetics , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Cell Differentiation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Neoplasms, Basal Cell/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Stem Cells/metabolism , Stem Cells/pathology
8.
Proc Natl Acad Sci U S A ; 111(35): 12787-92, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25139989

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3), a latent transcription factor associated with inflammatory signaling and innate and adaptive immune responses, is known to be aberrantly activated in a wide variety of cancers. In vitro analysis of STAT3 in human cancer cell lines has elucidated a number of specific targets associated with poor prognosis in breast cancer. However, to date, no comparison of cancer subtype and gene expression associated with STAT3 signaling in human patients has been reported. In silico analysis of human breast cancer microarray and reverse-phase protein array data was performed to identify expression patterns associated with STAT3 in basal-like and luminal breast cancers. Results indicate clearly identifiable STAT3-regulated signatures common to basal-like breast cancers but not to luminal A or luminal B cancers. Furthermore, these differentially expressed genes are associated with immune signaling and inflammation, a known phenotype of basal-like cancers. These findings demonstrate a distinct role for STAT3 signaling in basal breast cancers, and underscore the importance of considering subtype-specific molecular pathways that contribute to tissue-specific cancers.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasms, Basal Cell/genetics , STAT3 Transcription Factor/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Computational Biology , Female , Gene Expression Profiling , Humans , MAP Kinase Signaling System/physiology , MicroRNAs/genetics , Microarray Analysis , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/pathology , Phosphorylation/physiology , RNA, Messenger/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , STAT3 Transcription Factor/metabolism
9.
J Biol Chem ; 290(31): 19173-83, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26070560

ABSTRACT

FOXF2 (forkhead box F2) is a mesenchyme-specific transcription factor that plays a critical role in tissue homeostasis through the maintenance of epithelial polarity. In a previous study, we demonstrated that FOXF2 is specifically expressed in basal-like breast cancer (BLBC) cells and functions as an epithelial-mesenchymal transition suppressor. FOXF2 deficiency enhances the metastatic ability of BLBC cells through activation of the epithelial-mesenchymal transition program, but reduces cell proliferation. In this study, we demonstrate that CpG island methylation of the FOXF2 proximal promoter region is involved in the regulatory mechanism of the subtype-specific expression of FOXF2 in breast cancer cells. DNMT1, DNMT3A, and DNMT3B commonly or individually contributed to this DNA methylation in different breast cancer cells. SP1 regulated the transcriptional activity of FOXF2 through direct binding to the proximal promoter region, whereas this binding was abrogated through DNA methylation. FOXF2 mediated the SP1-regulated suppression of progression and promotion of proliferation of non-methylated BLBC cells. Thus, we conclude that the subtype-specific expression and function of FOXF2 in breast cancer cells are regulated through the combined effects of DNA methylation and SP1 transcriptional regulation.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Forkhead Transcription Factors/metabolism , Neoplasms, Basal Cell/genetics , Sp1 Transcription Factor/physiology , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Movement , Cell Proliferation , CpG Islands , Disease-Free Survival , Epigenesis, Genetic , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Molecular Sequence Data , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/mortality , Promoter Regions, Genetic , Up-Regulation
10.
BMC Cancer ; 16: 191, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26951085

ABSTRACT

BACKGROUND: Group IVA cytosolic phospholipase A2 (cPLA2α) plays an important role in tumorigenesis and angiogenesis. It is overexpressed in basal-like breast cancer (BLBC), which is aggressive and usually triple-negative, making it unresponsive to current targeted therapies. Here, we evaluated the anti-angiogenic effects of a specific cPLA2α inhibitor, AVX235, in a patient-derived triple-negative BLBC model. METHODS: Mice bearing orthotopic xenografts received i.p. injections of AVX235 or DMSO vehicle daily for 1 week and then every other day for up to 19 days. Six treated and six control mice were terminated after 2 days of treatment, and the tumors excised for high resolution magic angle spinning magnetic resonance spectroscopy (HR MAS MRS) and prostaglandin E2 (PGE2) enzyme immunoassay (EIA) analysis. A 1-week imaging study was performed on a separate cohort of mice. Longitudinal dynamic contrast enhanced (DCE)-MRI was performed before, after 4 days, and after 1 week of treatment. The mice were then perfused with a radiopaque vascular casting agent, and the tumors excised for micro-CT angiography. Subsequently, tumors were sectioned and stained with lectin and for Ki67 or α-smooth muscle actin to quantify endothelial cell proliferation and vessel maturity, respectively. Partial least squares discriminant analysis was performed on the multivariate HR MAS MRS data, and non-parametric univariate analyses using Mann-Whitney U tests (α = 0.05) were performed on all other data. RESULTS: Glycerophosphocholine and PGE2 levels, measured by HR MAS MRS and EIA, respectively, were lower in treated tumors after 2 days of treatment. These molecular changes are expected downstream effects of cPLA2α inhibition and were followed by significant tumor growth inhibition after 8 days of treatment. DCE-MRI revealed that AVX235 treatment caused a decrease in tumor perfusion. Concordantly, micro-CT angiography showed that vessel volume fraction, density, and caliber were reduced in treated tumors. Moreover, histology showed decreased endothelial cell proliferation and fewer immature vessels in treated tumors. CONCLUSIONS: These results demonstrate that cPLA2α inhibition with AVX235 resulted in decreased vascularization and perfusion and subsequent inhibition of tumor growth. Thus, cPLA2α inhibition may be a potential new therapeutic option for triple-negative basal-like breast cancer.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Neoplasms, Basal Cell/pathology , Neovascularization, Pathologic , Phospholipase A2 Inhibitors/pharmacology , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Dinoprostone/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Magnetic Resonance Imaging , Mice , Neoplasms, Basal Cell/drug therapy , Neoplasms, Basal Cell/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Phospholipases A2, Cytosolic/metabolism , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Tumor Burden/drug effects , X-Ray Microtomography , Xenograft Model Antitumor Assays
11.
Breast Cancer Res Treat ; 152(2): 347-56, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26109346

ABSTRACT

A large number of DNA copy number alterations (CNAs) exist in human breast cancers, and thus characterizing the most frequent CNAs is key to advancing therapeutics because it is likely that these regions contain breast tumor 'drivers' (i.e., cancer causal genes). This study aims to characterize the genomic landscape of breast cancer CNAs and identify potential subtype-specific drivers using a large set of human breast tumors and genetically engineered mouse (GEM) mammary tumors. Using a novel method called SWITCHplus, we identified subtype-specific DNA CNAs occurring at a 15% or greater frequency, which excluded many well-known breast cancer-related drivers such as amplification of ERBB2, and deletions of TP53 and RB1. A comparison of CNAs between mouse and human breast tumors identified regions with shared subtype-specific CNAs. Additional criteria that included gene expression-to-copy number correlation, a DawnRank network analysis, and RNA interference functional studies highlighted candidate driver genes that fulfilled these multiple criteria. Numerous regions of shared CNAs were observed between human breast tumors and GEM mammary tumor models that shared similar gene expression features. Specifically, we identified chromosome 1q21-23 as a Basal-like subtype-enriched region with multiple potential driver genes including PI4KB, SHC1, and NCSTN. This step-wise computational approach based on a cross-species comparison is applicable to any tumor type for which sufficient human and model system DNA copy number data exist, and in this instance, highlights that a single region of amplification may in fact harbor multiple driver genes.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Chromosome Mapping , Chromosomes, Human, Pair 1 , Oncogenes , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Computational Biology , DNA Copy Number Variations , Databases, Nucleic Acid , Female , Gene Dosage , Gene Regulatory Networks , Humans , Mice , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/pathology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Species Specificity
12.
Ultrastruct Pathol ; 39(5): 306-17, 2015.
Article in English | MEDLINE | ID: mdl-26107428

ABSTRACT

Aquaporin-3 (AQP3), is an aquaglyceroporin, that plays a role in cell proliferation, tumorigenesis, and cell migration. This study aimed at evaluating the possible role of AQP3 in nonmelanoma skin cancer (NMSC) pathogenesis through its immunohistochemical expression in skin biopsies of these diseases. One-hundred and thirty cutaneous specimens were studied. These included 60 cases of NMSC and 40 normal skin and 30 psoriasis samples, from age- and gender-matched subjects, as a control group. AQP3 was expressed in 66.7% of basal cell carcinoma (BCC) cases and in 93.3% of squamous cell carcinoma (SCC) cases. Higher AQP3 expression (p = .01), expression percentage (p = .01), and H score (p = .04) were significantly associated with SCC compared to BCC. Normal skin and psoriasis showed significantly higher AQP3 expression (p = .001, p < .001, respectively), expression percentage (p < .001 for both), and H score values (p < .001, p = .001, respectively) compared to NMSC. Higher H score values in BCC were significantly associated with female gender (p = .02) and with nodular lesions (p > .001). Higher H score values in SCC were significantly associated with grade III tumors (p = .04) and AQP3 percentage of expression was significantly correlated with grade III tumors (r = .48, p = .009). In conclusion, AQP3 may play a role in NMSC pathogenesis. This probably occurs through aquaporin-mediated glycerol transport and ATP generation. Its downregulation, observed in the current work, is mostly a result of excessive proliferation. Further studies are needed to investigate the therapeutic effect of its inhibition in NMSC treatment.


Subject(s)
Aquaporin 3/biosynthesis , Carcinoma, Squamous Cell/metabolism , Neoplasms, Basal Cell/metabolism , Skin Neoplasms/metabolism , Adult , Aged , Aquaporin 3/analysis , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasms, Basal Cell/pathology , Skin Neoplasms/pathology
13.
Breast Cancer Res ; 16(4): R78, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25069832

ABSTRACT

INTRODUCTION: In breast cancer, distinct expression profiles of microRNAs (miRNAs) have been associated with molecular subgroups and clinicopathological characteristics, implicating a diagnostic and prognostic role of miRNAs. However, the biological functions of deregulated miRNAs in tumor progression are not yet completely defined. In this study, we investigated the function of miR-18a in regulating breast cancer metastasis through the hypoxia-inducible factor 1α (HIF1A)-dependent hypoxic response. METHODS: An orthotopic metastatic breast cancer xenograft model (MDA-MB-231 cells) was used to identify miRNAs associated with spontaneous lung metastasis. The function of miR-18a in regulating HIF1A expression, as well as cellular responses to hypoxia and metastasis, were then studied in vitro and in vivo by assessing ectopic miR-18a expression or miR-18a inhibition. miRNA-mRNA interactions (AGO2 immunoprecipitation and 3' untranslated region Luciferase reporter assays), gene expression (quantitative PCR and microarray), cell migration and invasion, and cell growth were assessed under normoxic or hypoxic conditions, complemented by orthotopic xenograft of tumor cells to the mammary fat pad to investigate the effect of modulating miR-18a expression on primary tumor growth and lung metastasis. Last, clinically relevant correlations between miR-18a, HIF1A, hypoxia-responsive gene expression and distant metastasis-free survival (DMFS) were assessed using published expression array breast tumors data sets. RESULTS: miRNAs encoded by the MIR17HG gene were downregulated in lung metastases compared to primary tumors. Ectopic expression of miR-18a, a MIR17HG family member, in a metastatic variant of MDA-MB-231 cells reduced primary tumor growth and lung metastasis, whereas miR-18a inhibition in the parental cells promoted tumor growth and lung metastasis. We identified HIF1A as a direct target of miR-18a. Modulating miR-18a expression significantly affected hypoxic gene expression, cell invasiveness and sensitivity to anoikis and hypoxia in vitro in a HIF1A-dependent manner. Analysis of previously published data revealed that higher expression of HIF1A and a panel of hypoxic genes is associated with shorter DMFS interval in patients with basal-like breast tumors, and that, within this subtype, miR-18a expression is inversely correlated with hypoxic gene expression. Together, these data support a role of miR-18a in repressing distant metastasis through a HIF1A-dependent pathway. CONCLUSIONS: The results of this study reveal a novel role for miR-18a in targeting HIF1A and repressing metastasis of basal-like breast tumors.


Subject(s)
Breast Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/metabolism , MicroRNAs/physiology , Neoplasms, Basal Cell/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Hypoxia , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/secondary , MCF-7 Cells , Mice, Inbred NOD , Mice, SCID , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/secondary , RNA Interference
14.
Int J Cancer ; 134(10): 2424-36, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24615768

ABSTRACT

MET, a cell surface receptor for hepatocyte growth factor, is involved in the development of triple-negative/basal-like breast cancer (TNBC/BLBC). However, its utility as a therapeutic target in this subtype of breast cancer is poorly understood. To evaluate MET fully as a potential therapeutic target for TNBC/BLBC, we investigated the relationship between MET expression and clinical outcomes of patients with breast cancer and the functional effect of MET inhibition. Using automated immunohistochemistry (Ventana), we analyzed MET expression in 924 breast cancer patients with relevant clinicopathologic parameters. BLBC showed the strongest relationship with MET expression (57.5%, p < 0.001). High expression of MET in breast cancer resulted in poor overall survival (p = 0.001) and disease-free survival (DFS, p = 0.010). MET expression was relatively high in TNBC cell lines, and the silencing of MET via small interfering RNA reduced cell proliferation and migration. We observed reduced TNBC cell viability after treatment with the MET inhibitor PHA-665752. In the most drug-resistant cell line, MDA-MB-468, which showed elevated epidermal growth factor receptor (EGFR) expression, silencing of EGFR resulted in increased sensitivity to PHA-665752 treatment. We confirmed that PHA-665752 synergizes with the EGFR inhibitor erlotinib to decrease the viability of MDA-MB-468 cells. TNBC patients coexpressing MET and EGFR showed significantly worse DFS than that in patients expressing EGFR alone (p = 0.021). Our findings strongly suggest that MET may be a therapeutic target in TNBC and that the combined therapy targeting MET and EGFR may be beneficial for the treatment of TNBC/BLBC patients.


Subject(s)
Breast Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Molecular Targeted Therapy/methods , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , Immunohistochemistry , Indoles/pharmacology , Kaplan-Meier Estimate , Middle Aged , Neoplasms, Basal Cell/drug therapy , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Quinazolines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sulfones/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
15.
Br J Cancer ; 111(2): 326-38, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24901235

ABSTRACT

BACKGROUND: The multi-exon CSPP1 gene, encoding for centrosome and microtubule-associated proteins involved in ciliogenesis and cell division, is a candidate oncogene in luminal breast cancer but expression of CSPP1 proteins remained unexplored. METHODS: CSPP1 gene and protein expression was examined in normal mammary tissue, human breast cancer cell lines, and primary breast cancer biopsies from two patient cohorts. Cell type and epitope-dependent subcellular-specific CSPP1 staining pattern in normal mammary gland epithelium and cancer biopsies were correlated to molecular and clinical parameters. RESULTS: A novel, nuclear localised CSPP1 isoform was exclusively detected in luminal epithelial cells, whereas cytoplasmic CSPP-L was generally expressed in normal mammary epithelium. Luminal cell-related nuclear CSPP1 expression was preserved in type-matched cell lines and carcinomas, and correlated to gene copy number and mRNA expression. In contrast, basal-like carcinomas displayed generally lower CSPP1 mRNA expression. Yet, a subgroup of basal-like breast carcinomas depicted nuclear CSPP1 expression, displayed luminal traits, and differed from nuclear CSPP1 devoid counterparts in expression of eight genes. Eight-gene signature defined groups of basal-like tumours from an independent cohort showed significant differences in survival. CONCLUSIONS: Differential expression of a nuclear CSPP1 isoform identified biologically and clinically distinct subgroups of basal-like breast carcinoma.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins/biosynthesis , Microtubule-Associated Proteins/biosynthesis , Neoplasms, Basal Cell/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression , Gene Expression Profiling , HEK293 Cells , Humans , Immunohistochemistry , MCF-7 Cells , Microtubule-Associated Proteins/genetics , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/pathology , Transfection
16.
Mol Carcinog ; 53(10): 841-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23625632

ABSTRACT

Lung adenocarcinoma (AdC) and lung squamous cell carcinoma (SCC) are the most common non-small cell lung cancer (NSCLC) subtypes, however, most genetic mouse models of lung cancer produce predominantly, if not exclusively, AdC. Whether this is secondary to targeting mutations to the distal airway cells or to the use of activating Kras mutations that drive AdC formation is unknown. We previously showed that targeting Kras(G12D) activation and transforming growth factor ß receptor type II (TGFßRII) deletion to airway basal cells via a keratin promoter induced formation of both lung AdC and SCC. In this study we assessed if targeting phosphatase and tensin homologue (PTEN) deletion to airway basal cells could initiate lung tumor formation or increase lung SCC formation. We found that PTEN deletion is capable of initiating both lung AdC and SCC formation when targeted to basal cells and although PTEN deletion is a weaker tumor initiator than Kras(G12D) with low tumor multiplicity and long latency, tumors initiated by PTEN deletion were larger and displayed more malignant conversion than Kras(G12D) initiated tumors. That PTEN deletion did not increase lung SCC formation compared to Kras(G12D) activation, suggests that the initiating genetic event does not dictate tumor histology when genetic alterations are targeted to a specific cell. These studies also confirm that basal cells of the conducting airway are capable of giving rise to multiple NSCLC tumor types.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Neoplasms, Basal Cell/metabolism , PTEN Phosphohydrolase/genetics , Animals , Carcinoma, Squamous Cell/genetics , Gene Deletion , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Neoplasms, Basal Cell/genetics , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins p21(ras)/metabolism
17.
Mol Cell Proteomics ; 11(2): M111.011932, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086602

ABSTRACT

RNA helicase DDX5 (also p68) is involved in all aspects of RNA metabolism and serves as a transcriptional coregulator, but its functional role in breast cancer remains elusive. Here, we report an integrative biology study of DDX5 in breast cancer, encompassing quantitative proteomics, global MicroRNA profiling, and detailed biochemical characterization of cell lines and human tissues. We showed that protein expression of DDX5 increased progressively from the luminal to basal breast cancer cell lines, and correlated positively with that of CD44 in the basal subtypes. Through immunohistochemistry analyses of tissue microarrays containing over 200 invasive human ductal carcinomas, we observed that DDX5 was up-regulated in the majority of malignant tissues, and its expression correlated strongly with those of Ki67 and EGFR in the triple-negative tumors. We demonstrated that DDX5 regulated a subset of MicroRNAs including miR-21 and miR-182 in basal breast cancer cells. Knockdown of DDX5 resulted in reorganization of actin cytoskeleton and reduction of cellular proliferation. The effects were accompanied by up-regulation of tumor suppressor PDCD4 (a known miR-21 target); as well as up-regulation of cofilin and profilin, two key proteins involved in actin polymerization and cytoskeleton maintenance, as a consequence of miR-182 down-regulation. Treatment with miR-182 inhibitors resulted in morphologic phenotypes resembling those induced by DDX5 knockdown. Using bioinformatics tools for pathway and network analyses, we confirmed that the network for regulation of actin cytoskeleton was predominantly enriched for the predicted downstream targets of miR-182. Our results reveal a new functional role of DDX5 in breast cancer via the DDX5→miR-182→actin cytoskeleton pathway, and suggest the potential clinical utility of DDX5 and its downstream MicroRNAs in the theranostics of breast cancer.


Subject(s)
Actins/metabolism , Breast Neoplasms/genetics , Cytoskeleton/physiology , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms, Basal Cell/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Profiling , Humans , Immunoenzyme Techniques , MicroRNAs/metabolism , Neoplasm Invasiveness , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/pathology , Oligonucleotide Array Sequence Analysis , Proteome/analysis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Array Analysis , Tumor Cells, Cultured
18.
Breast Cancer Res ; 15(5): R76, 2013.
Article in English | MEDLINE | ID: mdl-24008056

ABSTRACT

INTRODUCTION: Mammographic density is a strong risk factor for breast cancer overall, but few studies have examined the association between mammographic density and specific subtypes of breast cancer, especially aggressive basal-like breast cancers. Because basal-like breast cancers are less frequently screen-detected, it is important to understand how mammographic density relates to risk of basal-like breast cancer. METHODS: We estimated associations between mammographic density and breast cancer risk according to breast cancer subtype. Cases and controls were participants in the Carolina Breast Cancer Study (CBCS) who also had mammograms recorded in the Carolina Mammography Registry (CMR). A total of 491 cases had mammograms within five years prior to and one year after diagnosis and 528 controls had screening or diagnostic mammograms close to the dates of selection into CBCS. Mammographic density was reported to the CMR using Breast Imaging Reporting and Data System categories. The expression of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 1 and 2 (HER1 and HER2), and cytokeratin 5/6 (CK5/6) were assessed by immunohistochemistry and dichotomized as positive or negative, with ER+ and/or PR+, and HER2- tumors classified as luminal A and ER-, PR-, HER2-, HER1+ and/or CK5/6+ tumors classified as basal-like breast cancer. Triple negative tumors were defined as negative for ER, PR and HER2. Of the 491 cases 175 were missing information on subtypes; the remaining cases included 181 luminal A, 17 luminal B, 48 basal-like, 29 ER-/PR-/HER2+, and 41 unclassified subtypes. Odds ratios comparing each subtype to all controls and case-case odds ratios comparing mammographic density distributions in basal-like to luminal A breast cancers were estimated using logistic regression. RESULTS: Mammographic density was associated with increased risk of both luminal A and basal-like breast cancers, although estimates were imprecise. The magnitude of the odds ratio associated with mammographic density was not substantially different between basal-like and luminal A cancers in case­control analyses and case-case analyses (case-case OR = 1.08 (95% confidence interval: 0.30, 3.84)). CONCLUSIONS: These results suggest that risk estimates associated with mammographic density are not distinct for separate breast cancer subtypes (basal-like/triple negative vs. luminal A breast cancers). Studies with a larger number of basal-like breast cancers are needed to confirm our findings.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Mammary Glands, Human/abnormalities , Neoplasms, Basal Cell/metabolism , Neoplasms, Basal Cell/pathology , Adult , Aged , Biomarkers, Tumor , Breast Density , Case-Control Studies , Female , Humans , Immunohistochemistry , Middle Aged , Odds Ratio , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Registries , Risk
19.
Mol Cancer ; 12(1): 139, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24238140

ABSTRACT

BACKGROUND AND METHODS: Stem or progenitor cells from healthy tissues have the capacity to co-segregate their template DNA strands during mitosis. Here, we set out to test whether breast cancer cell lines also possess the ability to asymmetrically segregate their template DNA strands via non-random chromosome co-segregation, and whether this ability correlates with certain properties attributed to breast cancer stem cells (CSCs). We quantified the frequency of asymmetric segregation of template DNA strands in 12 human breast cancer cell lines, and correlated the frequency to molecular subtype, CD44+/CD24-/lo phenotype, and invasion/migration ability. We tested if co-culture with human mesenchymal stem cells, which are known to increase self-renewal, can alter the frequency of asymmetric segregation of template DNA in breast cancer. RESULTS: We found a positive correlation between asymmetric segregation of template DNA and the breast cancer basal-like and claudin-low subtypes. There was an inverse correlation between asymmetric segregation of template DNA and Her2 expression. Breast cancer samples with evidence of asymmetric segregation of template DNA had significantly increased invasion and borderline significantly increased migration abilities. Samples with high CD44+/CD24-/lo surface expression were more likely to harbor a consistent population of cells that asymmetrically segregated its template DNA; however, symmetric self-renewal was enriched in the CD44+/CD24-/lo population. Co-culturing breast cancer cells with human mesenchymal stem cells expanded the breast CSC pool and decreased the frequency of asymmetric segregation of template DNA. CONCLUSIONS: Breast cancer cells within the basal-like subtype can asymmetrically segregate their template DNA strands through non-random chromosome segregation. The frequency of asymmetric segregation of template DNA can be modulated by external factors that influence expansion or self-renewal of CSC populations. Future studies to uncover the underlying mechanisms driving asymmetric segregation of template DNA and dictating cell fate at the time of cell division may explain how CSCs are maintained in tumors.


Subject(s)
Breast Neoplasms/pathology , Chromosome Segregation , DNA/genetics , Neoplasms, Basal Cell/pathology , Neoplastic Stem Cells/physiology , Breast Neoplasms/metabolism , CD24 Antigen/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , DNA/metabolism , Female , Humans , Hyaluronan Receptors/metabolism , Mesenchymal Stem Cells/physiology , Neoplasm Invasiveness , Neoplasms, Basal Cell/metabolism , Phenotype
20.
Breast Cancer Res Treat ; 138(1): 81-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23412770

ABSTRACT

In epithelial-to-mesenchymal transition (EMT) epithelial cancer cells achieve mesenchymal features, essentially helping them to metastasize. There is some evidence that EMT could be increased in triple-negative (TNBC) or basal-like breast cancers, although more precise mechanisms considering e.g. EMT-regulating transcription factors are largely unknown. We assessed immunohistochemically vimentin (separately in in situ areas and in invasive cells) as an indicator of EMT, and also EMT-regulating transcription factors zeb1 (separately in stroma and tumour) and Sip1 (in nuclei and cytoplasm) in histological samples of 231 women with local or locally advanced invasive breast cancer. 51.1 % of patients had TNBC and 48.9 % oestrogen and progesterone receptor-positive and HER2 negative breast cancer. Basal-like breast cancers were defined as TNBC that also expressed epidermal growth factor receptor EGFR and/or cytokeratin 5/6. Vimentin expression in invasive cells was higher in TNBCs (p = 9 × 10(-12)) compared to non-TNBC tumours. Vimentin (p = 2 × 10(-6)), nuclear Sip1 (p = 0.035) and zeb1 in stroma (p = 0.013) were overexpressed in basal-like cancers compared to non-basal-like TNBCs. In non-TNBC group findings between studied markers and clinicopathological factors were rare. However, in TNBC cases, vimentin expression in invasive cells associated with poor differentiation (p = 0.00007), zeb1 expression in cancer cells with higher grade (p = 0.002), vascular invasion (p = 0.036) and larger T-class (p = 0.027), whereas stromal zeb1 associated with lymphatic vessel invasion (p = 0.036) and vascular invasion (p = 0.039). High nuclear Sip1 expression was prognostic for poor disease-free survival (p = 0.002) in the whole cohort. The current results emphasize the increased role of EMT in TNBC and especially in basal-like breast cancers. These observations also support the role of studied parameters in tumour progression.


Subject(s)
Breast Neoplasms/metabolism , Homeodomain Proteins/metabolism , Neoplasms, Basal Cell/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Up-Regulation , Vimentin/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease Progression , Female , Humans , Neoplasm Grading , Neoplasm Staging , Neoplasms, Basal Cell/mortality , Neoplasms, Basal Cell/pathology , Phenotype , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL