Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 11(9)2022 04 28.
Article in English | MEDLINE | ID: mdl-35563787

ABSTRACT

One of the major obstacles in treating brain cancers, particularly glioblastoma multiforme, is the occurrence of secondary tumor lesions that arise in areas of the brain and are inoperable while obtaining resistance to current therapeutic agents. Thus, gaining a better understanding of the cellular factors that regulate glioblastoma multiforme cellular movement is imperative. In our study, we demonstrate that the 5'-3' exoribonuclease XRN2 is important to the invasive nature of glioblastoma. A loss of XRN2 decreases cellular speed, displacement, and movement through a matrix of established glioblastoma multiforme cell lines. Additionally, a loss of XRN2 abolishes tumor formation in orthotopic mouse xenograft implanted with G55 glioblastoma multiforme cells. One reason for these observations is that loss of XRN2 disrupts the expression profile of several cellular factors that are important for tumor invasion in glioblastoma multiforme cells. Importantly, XRN2 mRNA and protein levels are elevated in glioblastoma multiforme patient samples. Elevation in XRN2 mRNA also correlates with poor overall patient survival. These data demonstrate that XRN2 is an important cellular factor regulating one of the major obstacles in treating glioblastomas and is a potential molecular target that can greatly enhance patient survival.


Subject(s)
Brain Neoplasms , Exoribonucleases , Glioblastoma , Animals , Brain Neoplasms/metabolism , Cell Movement/genetics , Cell Proliferation , Exoribonucleases/metabolism , Glioblastoma/metabolism , Humans , Mice , Neoplastic Processes , RNA, Messenger/therapeutic use
2.
Cancers (Basel) ; 13(13)2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34206916

ABSTRACT

Centromere Protein I (CENP-I) is a member of the CENP-H/I/K complex. CENP-H/I/K is a major component of the inner kinetochore and aids in ensuring proper chromosomal segregation during mitosis. In addition to this chromosomal segregation function, CENP-I also plays a role in DNA double-strand break (DSB) repair. Loss of CENP-I leads to increased endogenous 53BP1 foci and R-loop formation, while reducing cellular survival after ionizing radiation and Niraparib, a PARP1 small molecule inhibitor, exposures. Cells lacking CENP-I display delayed 53BP1 foci regression, an indication that DSB repair is impaired. Additionally, loss of CENP-I impairs the homologous recombination DSB repair pathway, while having no effect on the non-homologous end-joining pathway. Interestingly, we find that RNaseH1 expression restores HR capacity in CENP-I deficient cells. Importantly, CENP-I expression is elevated in glioma tissue as compared to normal brain tissue. This elevated expression also correlates with poor overall patient survival. These data highlight the multi-functional role CENP-I plays in maintaining genetic, as well as chromosomal, stability and tumor survival.

3.
Mol Cancer Ther ; 20(10): 1800-1808, 2021 10.
Article in English | MEDLINE | ID: mdl-34253593

ABSTRACT

Metastasis is the major cause of mortality in patients with breast cancer. Many signaling pathways have been linked to cancer invasiveness, but blockade of few protein components has succeeded in reducing metastasis. Thus, identification of proteins contributing to invasion that are manipulable by small molecules may be valuable in inhibiting spread of the disease. The protein kinase with no lysine (K) 1 (WNK1) has been suggested to induce migration of cells representing a range of cancer types. Analyses of mouse models and patient data have implicated WNK1 as one of a handful of genes uniquely linked to invasive breast cancer. Here, we present evidence that inhibition of WNK1 slows breast cancer metastasis. We show that depletion or inhibition of WNK1 reduces migration of several breast cancer cell lines in wound healing assays and decreases invasion in collagen matrices. Furthermore, WNK1 depletion suppresses expression of AXL, a tyrosine kinase implicated in metastasis. Finally, we demonstrate that WNK inhibition in mice attenuates tumor progression and metastatic burden. These data showing reduced migration, invasion, and metastasis upon WNK1 depletion in multiple breast cancer models suggest that WNK1 contributes to the metastatic phenotype, and that WNK1 inhibition may offer a therapeutic avenue for attenuating progression of invasive breast cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , WNK Lysine-Deficient Protein Kinase 1/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Imidazoles/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Pyrrolidines/pharmacology , Tumor Cells, Cultured , WNK Lysine-Deficient Protein Kinase 1/antagonists & inhibitors , WNK Lysine-Deficient Protein Kinase 1/genetics , Xenograft Model Antitumor Assays
4.
Int J Mol Sci ; 22(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374314

ABSTRACT

Using a data driven analysis of a high-content screen, we have uncovered new regulators of epithelial-to-mesenchymal transition (EMT) induced cell migration. Our results suggest that increased expression of miR614 can alter cell intrinsic gene expression to enhance single cell and collective migration in multiple contexts. Interestingly, miR614 specifically increased the expression of the EMT transcription factor Slug while not altering existing epithelial character or inducing other canonical EMT regulatory factors. Analysis of two different cell lines identified a set of genes whose expression is altered by the miR614 through direct and indirect mechanisms. Prioritization driven by functional testing of 25 of the miR614 suppressed genes uncovered the mitochondrial small GTPase Miro1 and the transmembrane protein TAPT1 as miR614 suppressed genes that inhibit migration. Notably, the suppression of either Miro1 or TAPT1 was sufficient to increase Slug expression and the rate of cell migration. Importantly, reduced TAPT1 expression correlated with an increased risk of relapse in breast cancer patients. Together, our results reveal how increased miR614 expression and the suppression of TAPT1 and Miro1 modulate the EMT state and migratory properties of breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , RNA, Neoplasm/biosynthesis , Breast Neoplasms/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Humans , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Neoplasm/genetics
5.
Cancers (Basel) ; 12(7)2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32645903

ABSTRACT

It was recently shown that the 5' to 3' exoribonuclease XRN2 is involved in the DNA damage response. Importantly, loss of XRN2 abrogates DNA double stranded break repair via the non-homologous end-joining pathway. However, the mechanistic details of how XRN2 functions in the non-homologous end-joining repair process are unknown. In this study, we elucidated that XRN2-mediated RNA:DNA hybrid resolution is required to allow Ku70 binding to DNA ends. These data suggest that XRN2 is required for the initiation of non-homologous end-joining repair. Interestingly, we uncovered a role for XRN2 in the homologous recombination repair pathway. Loss of XRN2 lead to a decrease in the repair of double strand breaks by homologous recombination. Strikingly, when we removed RNA:DNA hybrids by RNaseH1 over-expression, homologous recombination was not restored. We found RNA:DNA hybrid formation at and downstream of the DSB site, suggesting that unregulated transcription inhibits homologous recombination repair. In summary, our results indicate a relation between RNA:DNA hybrid resolution and double strand break repair pathway choice.

6.
Cancer Res ; 80(18): 3933-3944, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32661136

ABSTRACT

Defining how interactions between tumor subpopulations contribute to invasion is essential for understanding how tumors metastasize. Here, we find that the heterogeneous expression of the transcription factor ΔNp63 confers distinct proliferative and invasive epithelial-to-mesenchymal transition (EMT) states in subpopulations that establish a leader-follower relationship to collectively invade. A ΔNp63-high EMT program coupled the ability to proliferate with an IL1α- and miR-205-dependent suppression of cellular protrusions that are required to initiate collective invasion. An alternative ΔNp63-low EMT program conferred cells with the ability to initiate and lead collective invasion. However, this ΔNp63-low EMT state triggered a collateral loss of fitness. Importantly, rare growth-suppressed ΔNp63-low EMT cells influenced tumor progression by leading the invasion of proliferative ΔNp63-high EMT cells in heterogeneous primary tumors. Thus, heterogeneous activation of distinct EMT programs promotes a mode of collective invasion that overcomes cell intrinsic phenotypic deficiencies to induce the dissemination of proliferative tumor cells. SIGNIFICANCE: These findings reveal how an interaction between cells in different EMT states confers properties that are not induced by either EMT program alone.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Surface Extensions , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/pathology , Female , Humans , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , RNA, Small Interfering/metabolism , Spheroids, Cellular , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
7.
Int J Mol Sci ; 21(12)2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32549188

ABSTRACT

Cellular survival is dependent on the efficient replication and transmission of genomic information. DNA damage can be introduced into the genome by several different methods, one being the act of DNA replication. Replication is a potent source of DNA damage and genomic instability, especially through the formation of DNA double strand breaks (DSBs). DNA polymerase alpha is responsible for replication initiation. One subunit of the DNA polymerase alpha replication machinery is POLA2. Given the connection between replication and genomic instability, we decided to examine the role of POLA2 in DSB repair, as little is known about this topic. We found that loss of POLA2 leads to an increase in spontaneous DSB formation. Loss of POLA2 also slows DSB repair kinetics after treatment with etoposide and inhibits both of the major double strand break repair pathways: non-homologous end-joining and homologous recombination. In addition, loss of POLA2 leads to increased sensitivity to ionizing radiation and PARP1 inhibition. Lastly, POLA2 expression is elevated in glioblastoma multiforme tumors and correlates with poor overall patient survival. These data demonstrate a role for POLA2 in DSB repair and resistance to genotoxic stress.


Subject(s)
Brain Neoplasms/genetics , DNA Polymerase I/genetics , Glioma/genetics , Up-Regulation , Brain Neoplasms/mortality , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioma/mortality , Humans , Indazoles/pharmacology , Piperidines/pharmacology , Radiation, Ionizing , Survival Analysis , Up-Regulation/drug effects , Up-Regulation/radiation effects
8.
PLoS Genet ; 12(7): e1006107, 2016 07.
Article in English | MEDLINE | ID: mdl-27437695

ABSTRACT

XRN2 is a 5'-3' exoribonuclease implicated in transcription termination. Here we demonstrate an unexpected role for XRN2 in the DNA damage response involving resolution of R-loop structures and prevention of DNA double-strand breaks (DSBs). We show that XRN2 undergoes DNA damage-inducible nuclear re-localization, co-localizing with 53BP1 and R loops, in a transcription and R-loop-dependent process. XRN2 loss leads to increased R loops, genomic instability, replication stress, DSBs and hypersensitivity of cells to various DNA damaging agents. We demonstrate that the DSBs that arise with XRN2 loss occur at transcriptional pause sites. XRN2-deficient cells also exhibited an R-loop- and transcription-dependent delay in DSB repair after ionizing radiation, suggesting a novel role for XRN2 in R-loop resolution, suppression of replication stress, and maintenance of genomic stability. Our study highlights the importance of regulating transcription-related activities as a critical component in maintaining genetic stability.


Subject(s)
DNA Damage , DNA Replication , Exoribonucleases/genetics , Exoribonucleases/physiology , Transcription, Genetic , Tumor Suppressor p53-Binding Protein 1/genetics , Antineoplastic Agents/chemistry , Cell Nucleus/metabolism , DNA Helicases , DNA Repair , Gene Expression Regulation, Neoplastic , Genomic Instability , Genomics , HeLa Cells , Humans , Microscopy, Fluorescence , Multifunctional Enzymes , Neoplasms/drug therapy , Neoplasms/genetics , Plasmids/metabolism , RNA Helicases/metabolism , RNA, Small Interfering/metabolism
9.
Oncotarget ; 7(19): 28592-611, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27081041

ABSTRACT

Tumor invasion can be induced by changes in gene expression that alter cell phenotype. The transcription factor ΔNp63α promotes basal-like breast cancer (BLBC) migration by inducing the expression of the mesenchymal genes Slug and Axl, which confers cells with a hybrid epithelial/mesenchymal state. However, the extent of the ΔNp63α regulated genes that support invasive behavior is not known. Here, using gene expression analysis, ChIP-seq, and functional testing, we find that ΔNp63α promotes BLBC motility by inducing the expression of the atypical cadherin FAT2, the vesicular binding protein SNCA, the carbonic anhydrase CA12, the lipid binding protein CPNE8 and the kinase NEK1, along with Slug and Axl. Notably, lung squamous cell carcinoma migration also required ΔNp63α dependent FAT2 and Slug expression, demonstrating that ΔNp63α promotes migration in multiple tumor types by inducing mesenchymal and non-mesenchymal genes. ΔNp63α activation of FAT2 and Slug influenced E-cadherin localization to cell-cell contacts, which can restrict spontaneous cell movement. Moreover, live-imaging of spheroids in organotypic culture demonstrated that ΔNp63α, FAT2 and Slug were essential for the extension of cellular protrusions that initiate collective invasion. Importantly, ΔNp63α is co-expressed with FAT2 and Slug in patient tumors and the elevated expression of ΔNp63α, FAT2 and Slug correlated with poor patient outcome. Together, these results reveal how ΔNp63α promotes cell migration by directly inducing the expression of a cohort of genes with distinct cellular functions and suggest that FAT2 is a new regulator of collective invasion that may influence patient outcome.


Subject(s)
Cadherins/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling/methods , Humans , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
10.
Oncotarget ; 7(12): 14708-26, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26895102

ABSTRACT

Genes that are normally biased towards expression in the testis are often induced in tumor cells. These gametogenic genes, known as cancer-testis antigens (CTAs), have been extenstively investigated as targets for immunotherapy. However, despite their frequent detection, the degree to which CTAs support neoplastic invasion is poorly understood. Here, we find that the CTA genes SPANX-A/C/D and CTAG2 are coordinately induced in breast cancer cells and regulate distinct features of invasive behavior. Our functional analysis revealed that CTAG2 interacts with Pericentrin at the centrosome and is necessary for directional migration. Conversely, SPANX-A/C/D interacts with Lamin A/C at the inner nuclear membrane and is required for the formation of actin-rich cellular protrusions that reorganize the extracellular matrix. Importantly, SPANX-A/C/D was required for breast cancer cells to spontaneously metastasize to the lung, demonstrating that CTA reactivation can be critical for invasion dependent phenotypes in vivo. Moreover, elevated SPANX-A/C/D expression in breast cancer patient tumors correlated with poor outcome. Together, our results suggest that distinct CTAs promote tumor progression by regulating complementary cellular functions that are integrated together to induce invasive behavior.


Subject(s)
Antigens, Neoplasm/metabolism , Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Apoptosis , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Cancer Res ; 75(18): 3925-35, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26292362

ABSTRACT

Cell identity signals influence the invasive capability of tumor cells, as demonstrated by the selection for programs of epithelial-to-mesenchymal transition (EMT) during malignant progression. Breast cancer cells retain canonical epithelial traits and invade collectively as cohesive groups of cells, but the signaling pathways critical to their invasive capabilities are still incompletely understood. Here we report that the transcription factor ΔNp63α drives the migration of basal-like breast cancer (BLBC) cells by inducing a hybrid mesenchymal/epithelial state. Through a combination of expression analysis and functional testing across multiple BLBC cell populations, we determined that ΔNp63α induces migration by elevating the expression of the EMT program components Slug and Axl. Interestingly, ΔNp63α also increased the expression of miR-205, which can silence ZEB1/2 to prevent the loss of epithelial character caused by EMT induction. In clinical specimens, co-expression of various elements of the ΔNp63α pathway confirmed its implication in motility signaling in BLBC. We observed that activation of the ΔNp63α pathway occurred during the transition from noninvasive ductal carcinoma in situ to invasive breast cancer. Notably, in an orthotopic tumor model, Slug expression was sufficient to induce collective invasion of E-cadherin-expressing BLBC cells. Together, our results illustrate how ΔNp63α can drive breast cancer cell invasion by selectively engaging promigratory components of the EMT program while, in parallel, still promoting the retention of epithelial character.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Breast Neoplasms/mortality , Cadherins/biosynthesis , Cadherins/genetics , Cell Movement , Disease Progression , Epithelial Cells/pathology , Female , Heterografts , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/biosynthesis , MicroRNAs/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured , Axl Receptor Tyrosine Kinase
12.
J Clin Invest ; 125(5): 1927-43, 2015 May.
Article in English | MEDLINE | ID: mdl-25844900

ABSTRACT

Tumor cells can engage in a process called collective invasion, in which cohesive groups of cells invade through interstitial tissue. Here, we identified an epigenetically distinct subpopulation of breast tumor cells that have an enhanced capacity to collectively invade. Analysis of spheroid invasion in an organotypic culture system revealed that these "trailblazer" cells are capable of initiating collective invasion and promote non-trailblazer cell invasion, indicating a commensal relationship among subpopulations within heterogenous tumors. Canonical mesenchymal markers were not sufficient to distinguish trailblazer cells from non-trailblazer cells, suggesting that defining the molecular underpinnings of the trailblazer phenotype could reveal collective invasion-specific mechanisms. Functional analysis determined that DOCK10, ITGA11, DAB2, PDFGRA, VASN, PPAP2B, and LPAR1 are highly expressed in trailblazer cells and required to initiate collective invasion, with DOCK10 essential for metastasis. In patients with triple-negative breast cancer, expression of these 7 genes correlated with poor outcome. Together, our results indicate that spontaneous conversion of the epigenetic state in a subpopulation of cells can promote a transition from in situ to invasive growth through induction of a cooperative form of collective invasion and suggest that therapeutic inhibition of trailblazer cell invasion may help prevent metastasis.


Subject(s)
Breast Neoplasms/pathology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Proteins/physiology , Animals , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Cell Surface Extensions/physiology , Cell Surface Extensions/ultrastructure , Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition , Extracellular Matrix , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/pathology , Neoplasm Proteins/genetics , RNA Interference , Specific Pathogen-Free Organisms , Spheroids, Cellular , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
13.
Cancer Res ; 71(21): 6857-66, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21908556

ABSTRACT

Most ductal breast carcinoma cells are weakly invasive in vitro and in vivo, suggesting that components of their microenvironment may facilitate a transition from in situ to invasive stages during progression. Here, we report that coculture of mammary fibroblasts specifically triggers invasive behavior in basal-type breast cancer cells through a ligand independent mechanism. When cultured alone in organotypic culture, both basal- and luminal-type breast cancer cells formed noninvasive spheroids with characteristics of ductal carcinoma in situ (DCIS). In contrast, when cocultured with mammary fibroblasts, basal-type spheroids exhibited invasive character whereas the luminal-type spheroids retained a benign and noninvasive duct-like architecture. Real-time imaging and functional studies revealed that the specificity of invasion was linked to a unique capacity of basal-type breast cancer cells to move within spheroids. Mammary fibroblasts induced invasion by triggering basal-type breast cancer cells to convert from a noninvasive program of mammary epithelial morphogenesis to an invasive program of sprouting endothelial angiogenesis. Contrary to the existing invasion models, soluble ligands produced by the fibroblasts were not sufficient to trigger invasion. Instead, basal-type invasion relied upon a Cdc42-dependent reorganization of collagen fibers in the extracellular matrix by fibroblasts. Inhibiting basal-type cell movement with clinically relevant drugs blocked invasion both in organotypic culture and in animals, suggesting a new treatment strategy for early-stage patients. Together our findings establish that fibroblast recruitment by basal-type breast cancer cells into early-stage tumors is sufficient to trigger their conversion from a benign, noninvasive DCIS-like stage to a malignant invasive stage. Furthermore, our findings suggest that different subtypes of breast cancer may require distinct types of contributions from the microenvironment to undergo malignant progression.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Neoplasm Invasiveness/physiopathology , Tumor Microenvironment/physiology , Animals , Breast/cytology , Breast Neoplasms/classification , Carcinoma, Ductal, Breast/classification , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured/cytology , Cells, Cultured/physiology , Coculture Techniques , Collagen/physiology , Culture Media, Conditioned/pharmacology , Extracellular Matrix/physiology , Extracellular Matrix/ultrastructure , Female , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Mice , Mice, SCID , Neoplasm Invasiveness/prevention & control , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/pharmacology , Spheroids, Cellular/physiology , Spheroids, Cellular/transplantation , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/physiology , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...