Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Dev Cell ; 57(24): 2714-2730.e8, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36538894

ABSTRACT

Cancer stem cells (CSCs) may serve as the cellular seeds of tumor recurrence and metastasis, and they can be generated via epithelial-mesenchymal transitions (EMTs). Isolating pure populations of CSCs is difficult because EMT programs generate multiple alternative cell states, and phenotypic plasticity permits frequent interconversions between these states. Here, we used cell-surface expression of integrin ß4 (ITGB4) to isolate highly enriched populations of human breast CSCs, and we identified the gene regulatory network operating in ITGB4+ CSCs. Specifically, we identified ΔNp63 and p73, the latter of which transactivates ΔNp63, as centrally important transcriptional regulators of quasi-mesenchymal CSCs that reside in an intermediate EMT state. We found that the transcriptional program controlled by ΔNp63 in CSCs is largely distinct from the one that it orchestrates in normal basal mammary stem cells and, instead, it more closely resembles a regenerative epithelial stem cell response to wounding. Moreover, quasi-mesenchymal CSCs repurpose this program to drive metastatic colonization via autocrine EGFR signaling.


Subject(s)
Mesenchymal Stem Cells , Neoplasms , Humans , Cell Line, Tumor , Neoplastic Stem Cells/metabolism , Signal Transduction , Epithelial-Mesenchymal Transition , Neoplasms/pathology
2.
J Cell Sci ; 134(6)2021 03 17.
Article in English | MEDLINE | ID: mdl-33097605

ABSTRACT

We report here the effects of targeted p120-catenin (encoded by CTNND1; hereafter denoted p120) knockout (KO) in a PyMT mouse model of invasive ductal (mammary) cancer (IDC). Mosaic p120 ablation had little effect on primary tumor growth but caused significant pro-metastatic alterations in the tumor microenvironment, ultimately leading to a marked increase in the number and size of pulmonary metastases. Surprisingly, although early effects of p120-ablation included decreased cell-cell adhesion and increased invasiveness, cells lacking p120 were almost entirely unable to colonized distant metastatic sites in vivo The relevance of this observation to human IDC was established by analysis of a large clinical dataset of 1126 IDCs. As reported by others, p120 downregulation in primary IDC predicted worse overall survival. However, as in the mice, distant metastases were almost invariably p120 positive, even in matched cases where the primary tumors were p120 negative. Collectively, our results demonstrate a strong positive role for p120 (and presumably E-cadherin) during metastatic colonization of distant sites. On the other hand, downregulation of p120 in the primary tumor enhanced metastatic dissemination indirectly via pro-metastatic conditioning of the tumor microenvironment.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/genetics , Cadherins/genetics , Catenins/genetics , Cell Adhesion , Female , Humans , Mice , Tumor Microenvironment , Delta Catenin
3.
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
4.
Cell ; 174(3): 716-729.e27, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29961576

ABSTRACT

Single-cell RNA sequencing technologies suffer from many sources of technical noise, including under-sampling of mRNA molecules, often termed "dropout," which can severely obscure important gene-gene relationships. To address this, we developed MAGIC (Markov affinity-based graph imputation of cells), a method that shares information across similar cells, via data diffusion, to denoise the cell count matrix and fill in missing transcripts. We validate MAGIC on several biological systems and find it effective at recovering gene-gene relationships and additional structures. Applied to the epithilial to mesenchymal transition, MAGIC reveals a phenotypic continuum, with the majority of cells residing in intermediate states that display stem-like signatures, and infers known and previously uncharacterized regulatory interactions, demonstrating that our approach can successfully uncover regulatory relations without perturbations.


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Algorithms , Cell Line , Epistasis, Genetic/genetics , Gene Regulatory Networks/genetics , Humans , Markov Chains , MicroRNAs/genetics , RNA, Messenger/genetics , Software
5.
Sci Transl Med ; 10(436)2018 04 11.
Article in English | MEDLINE | ID: mdl-29643230

ABSTRACT

Patients undergoing surgical resection of primary breast tumors confront a risk for metastatic recurrence that peaks sharply 12 to 18 months after surgery. The cause of early metastatic relapse in breast cancer has long been debated, with many ascribing these relapses to the natural progression of the disease. Others have proposed that some aspect of surgical tumor resection triggers the outgrowth of otherwise-dormant metastases, leading to the synchronous pattern of relapse. Clinical data cannot distinguish between these hypotheses, and previous experimental approaches have not provided clear answers. Such uncertainty hinders the development and application of therapeutic approaches that could potentially reduce early metastatic relapse. We describe an experimental model system that definitively links surgery and the subsequent wound-healing response to the outgrowth of tumor cells at distant anatomical sites. Specifically, we find that the systemic inflammatory response induced after surgery promotes the emergence of tumors whose growth was otherwise restricted by a tumor-specific T cell response. Furthermore, we demonstrate that perioperative anti-inflammatory treatment markedly reduces tumor outgrowth in this model, suggesting that similar approaches might substantially reduce early metastatic recurrence in breast cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Animals , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Mice , Neoplasm Metastasis/immunology , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism
6.
J Exp Med ; 214(8): 2243-2255, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28666979

ABSTRACT

Immunotherapy using checkpoint-blocking antibodies against targets such as CTLA-4 and PD-1 can cure melanoma and non-small cell lung cancer in a subset of patients. The presence of CD8 T cells in the tumor correlates with improved survival. We show that immuno-positron emission tomography (immuno-PET) can visualize tumors by detecting infiltrating lymphocytes and, through longitudinal observation of individual animals, distinguish responding tumors from those that do not respond to therapy. We used 89Zr-labeled PEGylated single-domain antibody fragments (VHHs) specific for CD8 to track the presence of intratumoral CD8+ T cells in the immunotherapy-susceptible B16 melanoma model in response to checkpoint blockade. A 89Zr-labeled PEGylated anti-CD8 VHH detected thymus and secondary lymphoid structures as well as intratumoral CD8 T cells. Animals that responded to CTLA-4 therapy showed a homogeneous distribution of the anti-CD8 PET signal throughout the tumor, whereas more heterogeneous infiltration of CD8 T cells correlated with faster tumor growth and worse responses. To support the validity of these observations, we used two different transplantable breast cancer models, yielding results that conformed with predictions based on the antimelanoma response. It may thus be possible to use immuno-PET and monitor antitumor immune responses as a prognostic tool to predict patient responses to checkpoint therapies.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , CTLA-4 Antigen/antagonists & inhibitors , Mammary Neoplasms, Experimental/therapy , Animals , CTLA-4 Antigen/physiology , Female , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/therapeutic use , Immunotherapy/methods , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/immunology , Mice , Neoplasm Transplantation , Positron-Emission Tomography/methods , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 114(12): E2337-E2346, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28270621

ABSTRACT

Neoplastic cells within individual carcinomas often exhibit considerable phenotypic heterogeneity in their epithelial versus mesenchymal-like cell states. Because carcinoma cells with mesenchymal features are often more resistant to therapy and may serve as a source of relapse, we sought to determine whether such cells could be further stratified into functionally distinct subtypes. Indeed, we find that a basal epithelial marker, integrin-ß4 (ITGB4), can be used to enable stratification of mesenchymal-like triple-negative breast cancer (TNBC) cells that differ from one another in their relative tumorigenic abilities. Notably, we demonstrate that ITGB4+ cancer stem cell (CSC)-enriched mesenchymal cells reside in an intermediate epithelial/mesenchymal phenotypic state. Among patients with TNBC who received chemotherapy, elevated ITGB4 expression was associated with a worse 5-year probability of relapse-free survival. Mechanistically, we find that the ZEB1 (zinc finger E-box binding homeobox 1) transcription factor activity in highly mesenchymal SUM159 TNBC cells can repress expression of the epithelial transcription factor TAp63α (tumor protein 63 isoform 1), a protein that promotes ITGB4 expression. In addition, we demonstrate that ZEB1 and ITGB4 are important in modulating the histopathological phenotypes of tumors derived from mesenchymal TNBC cells. Hence, mesenchymal carcinoma cell populations are internally heterogeneous, and ITGB4 is a mechanistically driven prognostic biomarker that can be used to identify the more aggressive subtypes of mesenchymal carcinoma cells in TNBC. The ability to rapidly isolate and mechanistically interrogate the CSC-enriched, partially mesenchymal carcinoma cells should further enable identification of novel therapeutic opportunities to improve the prognosis for high-risk patients with TNBC.


Subject(s)
Carcinoma/metabolism , Integrin beta4/metabolism , Mesoderm/metabolism , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/mortality , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Integrin beta4/genetics , Mesoderm/cytology , Prognosis , Transcription Factors/genetics , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
8.
Nature ; 543(7647): 681-686, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28329758

ABSTRACT

Post-mitotic, differentiated cells exhibit a variety of characteristics that contrast with those of actively growing neoplastic cells, such as the expression of cell-cycle inhibitors and differentiation factors. We hypothesized that the gene expression profiles of these differentiated cells could reveal the identities of genes that may function as tumour suppressors. Here we show, using in vitro and in vivo studies in mice and humans, that the mitochondrial protein LACTB potently inhibits the proliferation of breast cancer cells. Its mechanism of action involves alteration of mitochondrial lipid metabolism and differentiation of breast cancer cells. This is achieved, at least in part, through reduction of the levels of mitochondrial phosphatidylserine decarboxylase, which is involved in the synthesis of mitochondrial phosphatidylethanolamine. These observations uncover a novel mitochondrial tumour suppressor and demonstrate a connection between mitochondrial lipid metabolism and the differentiation program of breast cancer cells, thereby revealing a previously undescribed mechanism of tumour suppression.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation , Lipid Metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Tumor Suppressor Proteins/metabolism , beta-Lactamases/metabolism , Animals , Breast Neoplasms/genetics , Carboxy-Lyases/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Phosphatidylethanolamines/metabolism , Tumor Suppressor Proteins/genetics , beta-Lactamases/genetics
9.
Mol Cell ; 63(2): 277-292, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27373334

ABSTRACT

An abnormal differentiation state is common in BRCA1-deficient mammary epithelial cells, but the underlying mechanism is unclear. Here, we report a convergence between DNA repair and normal, cultured human mammary epithelial (HME) cell differentiation. Surprisingly, depleting BRCA1 or FANCD2 (Fanconi anemia [FA] proteins) or BRG1, a mSWI/SNF subunit, caused HME cells to undergo spontaneous epithelial-to-mesenchymal transition (EMT) and aberrant differentiation. This also occurred when wild-type HMEs were exposed to chemicals that generate DNA interstrand crosslinks (repaired by FA proteins), but not in response to double-strand breaks. Suppressed expression of ΔNP63 also occurred in each of these settings, an effect that links DNA damage to the aberrant differentiation outcome. Taken together with somatic breast cancer genome data, these results point to a breakdown in a BRCA/FA-mSWI/SNF-ΔNP63-mediated DNA repair and differentiation maintenance process in mammary epithelial cells that may contribute to sporadic breast cancer development.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/prevention & control , Cell Differentiation , DNA Damage , DNA Helicases/metabolism , DNA Repair , Epithelial Cells/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Mammary Glands, Human/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Acetaldehyde/pharmacology , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cisplatin/pharmacology , DNA Helicases/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Formaldehyde/pharmacology , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Mutation , Nuclear Proteins/genetics , Phenotype , RNA Interference , Signal Transduction , Transcription Factors/genetics , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Science ; 351(6277): aad3680, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26941323

ABSTRACT

The epithelial-to-mesenchymal transition enables carcinoma cells to acquire malignancy-associated traits and the properties of tumor-initiating cells (TICs). TICs have emerged in recent years as important targets for cancer therapy, owing to their ability to drive clinical relapse and enable metastasis. Here, we propose a strategy to eliminate mesenchymal TICs by inducing their conversion to more epithelial counterparts that have lost tumor-initiating ability. We report that increases in intracellular levels of the second messenger, adenosine 3',5'-monophosphate, and the subsequent activation of protein kinase A (PKA) induce a mesenchymal-to-epithelial transition (MET) in mesenchymal human mammary epithelial cells. PKA activation triggers epigenetic reprogramming of TICs by the histone demethylase PHF2, which promotes their differentiation and loss of tumor-initiating ability. This study provides proof-of-principle for inducing an MET as differentiation therapy for TICs and uncovers a role for PKA in enforcing and maintaining the epithelial state.


Subject(s)
Adenosine Monophosphate/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Second Messenger Systems , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Cholera Toxin/pharmacology , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation , Epigenesis, Genetic , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Histone Demethylases/metabolism , Homeodomain Proteins/metabolism , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mesoderm/drug effects , Mesoderm/metabolism , Mesoderm/pathology , Mice , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays
11.
Cell ; 158(5): 1094-1109, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171410

ABSTRACT

It is increasingly appreciated that oncogenic transformation alters cellular metabolism to facilitate cell proliferation, but less is known about the metabolic changes that promote cancer cell aggressiveness. Here, we analyzed metabolic gene expression in cancer cell lines and found that a set of high-grade carcinoma lines expressing mesenchymal markers share a unique 44 gene signature, designated the "mesenchymal metabolic signature" (MMS). A FACS-based shRNA screen identified several MMS genes as essential for the epithelial-mesenchymal transition (EMT), but not for cell proliferation. Dihydropyrimidine dehydrogenase (DPYD), a pyrimidine-degrading enzyme, was highly expressed upon EMT induction and was necessary for cells to acquire mesenchymal characteristics in vitro and for tumorigenic cells to extravasate into the mouse lung. This role of DPYD was mediated through its catalytic activity and enzymatic products, the dihydropyrimidines. Thus, we identify metabolic processes essential for the EMT, a program associated with the acquisition of metastatic and aggressive cancer cell traits.


Subject(s)
Epithelial-Mesenchymal Transition , Pyrimidines/metabolism , Animals , Carcinoma/metabolism , Cell Line, Tumor , Dihydrouracil Dehydrogenase (NADP)/genetics , Flow Cytometry , Gene Expression Profiling , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , RNA, Small Interfering/metabolism
12.
Cancer Cell ; 24(3): 347-64, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-24029232

ABSTRACT

The epithelial-mesenchymal transition program becomes activated during malignant progression and can enrich for cancer stem cells (CSCs). We report that inhibition of protein kinase C α (PKCα) specifically targets CSCs but has little effect on non-CSCs. The formation of CSCs from non-stem cells involves a shift from EGFR to PDGFR signaling and results in the PKCα-dependent activation of FRA1. We identified an AP-1 molecular switch in which c-FOS and FRA1 are preferentially utilized in non-CSCs and CSCs, respectively. PKCα and FRA1 expression is associated with the aggressive triple-negative breast cancers, and the depletion of FRA1 results in a mesenchymal-epithelial transition. Hence, identifying molecular features that shift between cell states can be exploited to target signaling components critical to CSCs.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Protein Kinase C-alpha/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism
13.
Cell Rep ; 3(5): 1430-9, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23623498

ABSTRACT

The adipocyte is central to organismal metabolism and exhibits significant functional and morphological plasticity during its formation and lifespan. Remarkable transformations of this cell occur during obesity and lactation, and thus it is essential to gain a better understanding of adipocyte function in these two metabolic processes. Considering the critical importance of the cellular organelle endoplasmic reticulum (ER) in adapting to fluctuations in synthetic processes, we explored the role of XBP1, a central regulator of ER adaptive responses, in adipocyte formation and function. Unexpectedly, deletion of adipocyte-XBP1 in vivo in mice (XBP1ΔAd) had no effect on adipocyte formation or on systemic homeostatic metabolism in mice fed a a regular or high-fat diet. However, during lactation, XBP1ΔAd dams displayed increased adiposity, decreased milk production, and decreased litter growth as compared with control dams. Moreover, we demonstrate that XBP1 is regulated during lactation and responds to prolactin to alter lipogenic gene expression. These results demonstrate a role for adipocyte-XBP1 in the regulation of lactational metabolism.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , 3T3-L1 Cells , Adiposity , Animals , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Diet, High-Fat , Endoplasmic Reticulum/metabolism , Female , Lactation/metabolism , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Prolactin/metabolism , RNA, Messenger/metabolism , RNA, Ribosomal, 18S/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors/deficiency , Transcription Factors/genetics , X-Box Binding Protein 1
14.
Development ; 139(10): 1754-64, 2012 May.
Article in English | MEDLINE | ID: mdl-22461563

ABSTRACT

Although p120-catenin (p120) is crucial for E-cadherin function, ablation experiments in epithelial tissues from different organ systems reveal markedly different effects. Here, we examine for the first time the consequences of p120 knockout during mouse mammary gland development. An MMTV-Cre driver was used to target knockout to the epithelium at the onset of puberty. p120 ablation was detected in approximately one-quarter of the nascent epithelium at the forth week post-partum. However, p120 null cells were essentially nonadherent, excluded from the process of terminal end bud (TEB) morphogenesis and lost altogether by week six. This elimination process caused a delay in TEB outgrowth, after which the gland developed normally from cells that had retained p120. Mechanistic studies in vitro indicate that TEB dysfunction is likely to stem from striking E-cadherin loss, failure of cell-cell adhesion and near total exclusion from the collective migration process. Our findings reveal an essential role for p120 in mammary morphogenesis.


Subject(s)
Catenins/metabolism , Mammary Glands, Animal/metabolism , Morphogenesis/physiology , Animals , Blotting, Western , Catenins/genetics , Cell Line , Cells, Cultured , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Humans , Immunohistochemistry , Mammary Glands, Animal/cytology , Mammary Glands, Animal/embryology , Mice , Morphogenesis/genetics , Wound Healing/genetics , Wound Healing/physiology , Delta Catenin
15.
J Clin Invest ; 121(10): 4106-17, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926469

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), one of the most lethal neoplasms, is characterized by an expanded stroma with marked fibrosis (desmoplasia). We previously generated pancreas epithelium-specific TGF-ß receptor type II (Tgfbr2) knockout mice in the context of Kras activation (mice referred to herein as Kras+Tgfbr2KO mice) and found that they developed aggressive PDAC that recapitulated the histological manifestations of the human disease. The mouse PDAC tissue showed strong expression of connective tissue growth factor (Ctgf), a profibrotic and tumor-promoting factor, especially in the tumor-stromal border area, suggesting an active tumor-stromal interaction. Here we show that the PDAC cells in Kras+Tgfbr2KO mice secreted much higher levels of several Cxc chemokines compared with mouse pancreatic intraepithelial neoplasia cells, which are preinvasive. The Cxc chemokines induced Ctgf expression in the pancreatic stromal fibroblasts, not in the PDAC cells themselves. Subcutaneous grafting studies revealed that the fibroblasts enhanced growth of PDAC cell allografts, which was attenuated by Cxcr2 inhibition. Moreover, treating the Kras+Tgfbr2KO mice with the CXCR2 inhibitor reduced tumor progression. The decreased tumor progression correlated with reduced Ctgf expression and angiogenesis and increased overall survival. Taken together, our data indicate that tumor-stromal interactions via a Cxcr2-dependent chemokine and Ctgf axis can regulate PDAC progression. Further, our results suggest that inhibiting tumor-stromal interactions might be a promising therapeutic strategy for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Pancreatic Neoplasms/therapy , Receptors, Interleukin-8B/antagonists & inhibitors , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/physiopathology , Chemokines, CXC/physiology , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/physiology , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/physiology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Interleukin-8B/physiology , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Stromal Cells/pathology , Stromal Cells/physiology , Tumor Microenvironment/physiology
16.
Proc Natl Acad Sci U S A ; 108(19): 7950-5, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21498687

ABSTRACT

Current models of stem cell biology assume that normal and neoplastic stem cells reside at the apices of hierarchies and differentiate into nonstem progeny in a unidirectional manner. Here we identify a subpopulation of basal-like human mammary epithelial cells that departs from that assumption, spontaneously dedifferentiating into stem-like cells. Moreover, oncogenic transformation enhances the spontaneous conversion, so that nonstem cancer cells give rise to cancer stem cell (CSC)-like cells in vitro and in vivo. We further show that the differentiation state of normal cells-of-origin is a strong determinant of posttransformation behavior. These findings demonstrate that normal and CSC-like cells can arise de novo from more differentiated cell types and that hierarchical models of mammary stem cell biology should encompass bidirectional interconversions between stem and nonstem compartments. The observed plasticity may allow derivation of patient-specific adult stem cells without genetic manipulation and holds important implications for therapeutic strategies to eradicate cancer.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Cell Dedifferentiation , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Breast/physiology , Breast Neoplasms/physiopathology , CD24 Antigen/metabolism , Cell Dedifferentiation/physiology , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Humans , Hyaluronan Receptors/metabolism , Mammary Glands, Animal/cytology , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Stem Cell Transplantation , Transplantation, Heterologous
17.
Cytokine Growth Factor Rev ; 21(1): 49-59, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20018551

ABSTRACT

The transforming growth factor beta (TGF-beta) has been studied with regard to the regulation of cell behavior for over three decades. A large body of research has been devoted to the regulation of epithelial cell and derivative carcinoma cell populations in vitro and in vivo. TGF-beta has been shown to inhibit epithelial cell cycle progression and promote apoptosis that together significantly contribute to the tumor suppressive role for TGF-beta during carcinoma initiation and progression. TGF-beta is also able to promote an epithelial to mesenchymal transition that has been associated with increased tumor cell motility, invasion and metastasis. However, it has now been shown that loss of carcinoma cell responsiveness to TGF-beta stimulation can also promote metastasis. Interestingly, enhanced metastasis in the absence of a carcinoma cell response to TGF-beta stimulation has been shown to involve increased chemokine production resulting in recruitment of pro-metastatic myeloid derived suppressor cell (MDSC) populations to the tumor microenvironment at the leading invasive edge. When present, MDSCs enhance angiogenesis, promote immune tolerance and provide matrix degrading enzymes that promote tumor progression and metastasis. Further, the recruitment of MDSC populations in this context likely enhances the classic role for TGF-beta in immune suppression since the MDSCs are an abundant source of TGF-beta production. Importantly, it is now clear that carcinoma-immune cell cross-talk initiated by TGF-beta signaling within the carcinoma cell is a significant determinant worth consideration when designing therapeutic strategies to manage tumor progression and metastasis.


Subject(s)
Inflammation/complications , Inflammation/physiopathology , Neoplasms/etiology , Neoplasms/physiopathology , Transforming Growth Factor beta/physiology , Animals , Epithelial Cells/physiology , Humans , Macrophages/physiology , Models, Biological , Monocytes/physiology , Myeloid Cells/physiology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/immunology , Signal Transduction
18.
Cell Cycle ; 8(20): 3319-27, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19806012

ABSTRACT

The transforming growth factor beta (TGFbeta) is a potent regulator of tumor initiation, progression and metastasis. It has been known for many years that TGFbeta signaling in the carcinoma cell can suppress or promote tumor progression depending on the context of stimulation. While the impact of TGFbeta on the carcinoma cell is significant, it is now generally accepted that primary and metastatic carcinoma progression is regulated by an intricate network of host-tumor cell interactions. Interestingly, recent results have revealed that gain or loss of TGFbeta signaling in carcinoma cells can promote metastasis through carcinoma cell derived TGFbeta dependent host-tumor cell interactions in vivo. Further, gain or complete abrogation of TGFbeta signaling was shown to result in gene expression signatures that correlated with poor patient prognosis in breast cancer. Specifically, the TGFbeta responsive gene expression signature correlated with poor prognosis for estrogen receptor negative (ER(-)) breast cancer while complete abrogation of TGFbeta signaling resulted in a correlation with poor outcome in lymph node positive (LN(+)) and ER(+) breast cancers. Importantly, in both cases the correlation with poor prognosis was linked to carcinoma cell derived interactions with the adjacent microenvironment. Together the current results suggest that, in addition to intrinsic carcinoma cell signaling, TGFbeta dependent host-tumor cell interactions should be considered when designing therapeutic strategies to manage carcinoma progression.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Transforming Growth Factor beta/metabolism , Breast Neoplasms/therapy , Carcinoma/secondary , Carcinoma/therapy , Chemokines/metabolism , Disease Progression , Female , Humans , Receptors, Estrogen/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics
19.
Mol Cancer Res ; 7(5): 645-53, 2009 May.
Article in English | MEDLINE | ID: mdl-19435821

ABSTRACT

A subset of neoplastic cells within human high-grade gliomas has features associated with stem cells. These cells may sustain glioma growth, and their stem-like properties may confer resistance to standard glioma treatments. Whether glioma stem cells derive from indigenous neural stem cells (NSC), or from tumor cells that have reacquired stem cell-like properties, is unknown. However, signaling pathways that are tightly regulated and central to NSC biology, including the Ras/Raf/Erk pathway, are hyperactive and pathogenic in gliomagenesis. Furthermore, data in animal models suggests that, in some cases, tumors are initiated in the subventricular zone (SVZ), a stem/progenitor cell niche in the mature brain. We activated oncogenic K-ras in mouse glioneuronal precursor cells and adult SVZ cells using GFAP-Cre. GFAP-Cre+/K-ras(G12D) mice showed a marked expansion of glial fibriallary acidic protein (GFAP)- and TUJ1-expressing cell populations in the SVZ. In addition, mice developed intermediate grade, infiltrating glioma with 100% penetrance. Tumors were consistently located in the amygdalohippocampal region and nearby cortex, often in association with the lateral ventricle and expanded SVZ. Tumor cells expressed markers associated with neural progenitor cells, including Olig2, Bmi-1, and PDGFR-alpha. These data suggest that infiltrating tumor cells may arise from NSC transformed by activation of oncogenic K-ras in vivo.


Subject(s)
Cerebral Ventricles/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glioma/metabolism , Integrases/metabolism , ras Proteins/metabolism , Animals , Cerebral Ventricles/pathology , Female , Glial Fibrillary Acidic Protein/genetics , Glioma/pathology , Immunohistochemistry , Integrases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , ras Proteins/genetics
20.
J Clin Invest ; 119(6): 1571-82, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19451693

ABSTRACT

In human breast cancer, loss of carcinoma cell-specific response to TGF-beta signaling has been linked to poor patient prognosis. However, the mechanisms through which TGF-beta regulates these processes remain largely unknown. In an effort to address this issue, we have now identified gene expression signatures associated with the TGF-beta signaling pathway in human mammary carcinoma cells. The results strongly suggest that TGF-beta signaling mediates intrinsic, stromal-epithelial, and host-tumor interactions during breast cancer progression, at least in part, by regulating basal and oncostatin M-induced CXCL1, CXCL5, and CCL20 chemokine expression. To determine the clinical relevance of our results, we queried our TGF-beta-associated gene expression signatures in 4 human breast cancer data sets containing a total of 1,319 gene expression profiles and associated clinical outcome data. The signature representing complete abrogation of TGF-beta signaling correlated with reduced relapse-free survival in all patients; however, the strongest association was observed in patients with estrogen receptor-positive (ER-positive) tumors, specifically within the luminal A subtype. Together, the results suggest that assessment of TGF-beta signaling pathway status may further stratify the prognosis of ER-positive patients and provide novel therapeutic approaches in the management of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokines/biosynthesis , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Antigens, Viral, Tumor/immunology , Breast Neoplasms/genetics , Cell Differentiation , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Polyomavirus/immunology , Prognosis , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Stem Cells/cytology , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...