Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
Cancer Res ; 81(3): 606-618, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32327406

ABSTRACT

Platelet-derived growth factor receptor-beta (PDGFRß) is a receptor tyrosine kinase found in cells of mesenchymal origin such as fibroblasts and pericytes. Activation of this receptor is dependent on paracrine ligand induction, and its preferred ligand PDGFB is released by neighboring epithelial and endothelial cells. While expression of both PDGFRß and PDGFB has been noted in patient breast tumors for decades, how PDGFB-to-PDGFRß tumor-stroma signaling mediates breast cancer initiation, progression, and metastasis remains unclear. Here we demonstrate this paracrine signaling pathway that mediates both primary tumor growth and metastasis, specifically, metastasis to the brain. Elevated levels of PDGFB accelerated orthotopic tumor growth and intracranial growth of mammary tumor cells, while mesenchymal-specific expression of an activating mutant PDGFRß (PDGFRßD849V) exerted proproliferative signals on adjacent mammary tumor cells. Stromal expression of PDGFRßD849V also promoted brain metastases of mammary tumor cells expressing high PDGFB when injected intravenously. In the brain, expression of PDGFRßD849V was observed within a subset of astrocytes, and aged mice expressing PDGFRßD849V exhibited reactive gliosis. Importantly, the PDGFR-specific inhibitor crenolanib significantly reduced intracranial growth of mammary tumor cells. In a tissue microarray comprised of 363 primary human breast tumors, high PDGFB protein expression was prognostic for brain metastases, but not metastases to other sites. Our results advocate the use of mice expressing PDGFRßD849V in their stromal cells as a preclinical model of breast cancer-associated brain metastases and support continued investigation into the clinical prognostic and therapeutic use of PDGFB-to-PDGFRß signaling in women with breast cancer. SIGNIFICANCE: These studies reveal a previously unknown role for PDGFB-to-PDGFRß paracrine signaling in the promotion of breast cancer brain metastases and support the prognostic and therapeutic clinical utility of this pathway for patients.See related article by Wyss and colleagues, p. 594.


Subject(s)
Breast Neoplasms , MicroRNAs , Animals , Brain/metabolism , Breast Neoplasms/genetics , Endothelial Cells/metabolism , Humans , Mice , Receptor, Platelet-Derived Growth Factor beta
3.
Oncogene ; 40(1): 189-202, 2021 01.
Article in English | MEDLINE | ID: mdl-33110236

ABSTRACT

Triple-negative breast cancer (TNBC) is the deadliest form of breast cancer. Unlike other types of breast cancer that can be effectively treated by targeted therapies, no such targeted therapy exists for all TNBC patients. The ADAR1 enzyme carries out A-to-I editing of RNA to prevent sensing of endogenous double-stranded RNAs. ADAR1 is highly expressed in breast cancer including TNBC. Here, we demonstrate that expression of ADAR1, specifically its p150 isoform, is required for the survival of TNBC cell lines. In TNBC cells, knockdown of ADAR1 attenuates proliferation and tumorigenesis. Moreover, ADAR1 knockdown leads to robust translational repression. ADAR1-dependent TNBC cell lines also exhibit elevated IFN stimulated gene expression. IFNAR1 reduction significantly rescued the proliferative defects of ADAR1 loss. These findings establish ADAR1 as a novel therapeutic target for TNBC tumors.


Subject(s)
Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Triple Negative Breast Neoplasms/pathology , Up-Regulation , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Neoplasm Transplantation , Protein Isoforms/metabolism , Receptor, Interferon alpha-beta/metabolism , Survival Analysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
4.
Cancer Res ; 80(19): 4172-4184, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32732220

ABSTRACT

Heterozygous mutations in the BRCA1 gene predispose women to breast and ovarian cancer, while biallelic BRCA1 mutations are a cause of Fanconi anemia (FA), a rare genetic disorder characterized by developmental abnormalities, early-onset bone marrow failure, increased risk of cancers, and hypersensitivity to DNA-crosslinking agents. BRCA1 is critical for homologous recombination of DNA double-strand breaks (DSB). Through its coiled-coil domain, BRCA1 interacts with an essential partner, PALB2, recruiting BRCA2 and RAD51 to sites of DNA damage. Missense mutations within the coiled-coil domain of BRCA1 (e.g., L1407P) that affect the interaction with PALB2 have been reported in familial breast cancer. We hypothesized that if PALB2 regulates or mediates BRCA1 tumor suppressor function, ablation of the BRCA1-PALB2 interaction may also elicit genomic instability and tumor susceptibility. We generated mice defective for the Brca1-Palb2 interaction (Brca1 L1363P in mice) and established MEF cells from these mice. Brca1 L1363P/L1363P MEF exhibited hypersensitivity to DNA-damaging agents and failed to recruit Rad51 to DSB. Brca1 L1363P/L1363P mice were viable but exhibited various FA symptoms including growth retardation, hyperpigmentation, skeletal abnormalities, and male/female infertility. Furthermore, all Brca1 L1363P/L1363P mice exhibited macrocytosis and died due to bone marrow failure or lymphoblastic lymphoma/leukemia with activating Notch1 mutations. These phenotypes closely recapitulate clinical features observed in patients with FA. Collectively, this model effectively demonstrates the significance of the BRCA1-PALB2 interaction in genome integrity and provides an FA model to investigate hematopoietic stem cells for mechanisms underlying progressive failure of hematopoiesis and associated development of leukemia/lymphoma, and other FA phenotypes. SIGNIFICANCE: A new Brca1 mouse model for Fanconi anemia (FA) complementation group S provides a system in which to study phenotypes observed in human FA patients including bone marrow failure.See related commentary by Her and Bunting, p. 4044.


Subject(s)
Breast Neoplasms , Fanconi Anemia , Animals , BRCA1 Protein/genetics , DNA Damage/genetics , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group N Protein/genetics , Female , Humans , Male , Mice , Phenotype , Tumor Suppressor Proteins/genetics
5.
Life Sci Alliance ; 1(5): e201800190, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30456390

ABSTRACT

The contribution of the tumor microenvironment to pancreatic ductal adenocarcinoma (PDAC) development is currently unclear. We therefore examined the consequences of disrupting paracrine Hedgehog (HH) signaling in PDAC stroma. Herein, we show that ablation of the key HH signaling gene Smoothened (Smo) in stromal fibroblasts led to increased proliferation of pancreatic tumor cells. Furthermore, Smo deletion resulted in proteasomal degradation of the tumor suppressor PTEN and activation of oncogenic protein kinase B (AKT) in fibroblasts. An unbiased proteomic screen identified RNF5 as a novel E3 ubiquitin ligase responsible for degradation of phosphatase and tensin homolog (PTEN) in Smo-null fibroblasts. Ring Finger Protein 5 (Rnf5) knockdown or pharmacological inhibition of glycogen synthase kinase 3ß (GSKß), the kinase that marks PTEN for ubiquitination, rescued PTEN levels and reversed the oncogenic phenotype, identifying a new node of PTEN regulation. In PDAC patients, low stromal PTEN correlated with reduced overall survival. Mechanistically, PTEN loss decreased hydraulic permeability of the extracellular matrix, which was reversed by hyaluronidase treatment. These results define non-cell autonomous tumor-promoting mechanisms activated by disruption of the HH/PTEN axis and identifies new targets for restoring stromal tumor-suppressive functions.

6.
Nat Commun ; 9(1): 2783, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30018330

ABSTRACT

The importance of the tumor-associated stroma in cancer progression is clear. However, it remains uncertain whether early events in the stroma are capable of initiating breast tumorigenesis. Here, we show that in the mammary glands of non-tumor bearing mice, stromal-specific phosphatase and tensin homolog (Pten) deletion invokes radiation-induced genomic instability in neighboring epithelium. In these animals, a single dose of whole-body radiation causes focal mammary lobuloalveolar hyperplasia through paracrine epidermal growth factor receptor (EGFR) activation, and EGFR inhibition abrogates these cellular changes. By analyzing human tissue, we discover that stromal PTEN is lost in a subset of normal breast samples obtained from reduction mammoplasty, and is predictive of recurrence in breast cancer patients. Combined, these data indicate that diagnostic or therapeutic chest radiation may predispose patients with decreased stromal PTEN expression to secondary breast cancer, and that prophylactic EGFR inhibition may reduce this risk.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/genetics , PTEN Phosphohydrolase/genetics , Radiation Tolerance/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Transformation, Neoplastic , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gamma Rays/adverse effects , Genomic Instability/drug effects , Genomic Instability/radiation effects , Humans , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/radiation effects , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/radiation effects , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/radiotherapy , Mice , PTEN Phosphohydrolase/deficiency , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/radiation effects
7.
Oncogene ; 37(32): 4428-4442, 2018 08.
Article in English | MEDLINE | ID: mdl-29720724

ABSTRACT

The chemokine CXCL12 has been shown to regulate breast tumor growth, however, its mechanism in initiating distant metastasis is not well understood. Here, we generated a novel conditional allele of Cxcl12 in mice and used a fibroblast-specific Cre transgene along with various mammary tumor models to evaluate CXCL12 function in the breast cancer metastasis. Ablation of CXCL12 in stromal fibroblasts of mice significantly delayed the time to tumor onset and inhibited distant metastasis in different mouse models. Elucidation of mechanisms using in vitro and in vivo model systems revealed that CXCL12 enhances tumor cell intravasation by increasing vascular permeability and expansion of a leaky tumor vasculature. Furthermore, our studies revealed CXCL12 enhances permeability by recruiting endothelial precursor cells and decreasing endothelial tight junction and adherence junction proteins. High expression of stromal CXCL12 in large cohort of breast cancer patients was directly correlated to blood vessel density and inversely correlated to recurrence and overall patient survival. In addition, our analysis revealed that stromal CXCL12 levels in combination with number of CD31+ blood vessels confers poorer patient survival compared to individual protein level. However, no correlation was observed between epithelial CXCL12 and patient survival or blood vessel density. Our findings describe the novel interactions between fibroblasts-derived CXCL12 and endothelial cells in facilitating tumor cell intrvasation, leading to distant metastasis. Overall, our studies indicate that cross-talk between fibroblast-derived CXCL12 and endothelial cells could be used as novel biomarker and strategy for developing tumor microenvironment based therapies against aggressive and metastatic breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CXCL12/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Neoplasm Invasiveness/pathology , Animals , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Mammary Neoplasms, Animal , Mice , Mice, Transgenic , Neoplasm Metastasis/pathology , Tumor Microenvironment/physiology
8.
Cancer Res ; 78(7): 1739-1750, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29339537

ABSTRACT

The initial molecular events that lead to malignant transformation of the fimbria of the fallopian tube (FT) through high-grade serous ovarian carcinoma (HGSC) remain poorly understood. In this study, we report that increased expression of signal transducer and activator of transcription 3 (pSTAT3 Tyr705) and suppression or loss of protein inhibitor of activated STAT3 (PIAS3) in FT likely drive HGSC. We evaluated human tissues-benign normal FT, tubal-peritoneal junction (TPJ), p53 signature FT tissue, tubal intraepithelial lesion in transition (TILT), serous tubal intraepithelial carcinoma (STIC) without ovarian cancer, and HGSC for expression of STAT3/PIAS3 (compared with their known TP53 signature) and their target proliferation genes. We observed constitutive activation of STAT3 and low levels or loss of PIAS3 in the TPJ, p53 signature, TILT, and STIC through advanced stage IV (HGSC) tissues. Elevated expression of pSTAT3 Tyr705 and decreased levels of PIAS3 appeared as early as TPJ and the trend continued until very advanced stage HGSC (compared with high PIAS3 and low pSTAT3 expression in normal benign FT). Exogenous expression of STAT3 in FT cells mediated translocation of pSTAT3 and c-Myc into the nucleus. In vivo experiments demonstrated that overexpression of STAT3 in FT secretory epithelial cells promoted tumor progression and metastasis, mimicking the clinical disease observed in patients with HGSC. Thus, we conclude that the STAT3 pathway plays a role in the development and progression of HGSC from its earliest premalignant states.Significance: Concomitant gain of pSTAT3 Tyr705 and loss of PIAS3 appear critical for initiation and development of high-grade serous carcinoma. Cancer Res; 78(7); 1739-50. ©2018 AACR.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Fallopian Tube Neoplasms/genetics , Molecular Chaperones/genetics , Ovarian Neoplasms/genetics , Precancerous Conditions/genetics , Protein Inhibitors of Activated STAT/genetics , STAT3 Transcription Factor/genetics , Animals , Cell Line, Tumor , Cell Movement , Cell Transformation, Neoplastic/genetics , Cystadenocarcinoma, Serous/pathology , Fallopian Tube Neoplasms/pathology , Fallopian Tubes/pathology , Female , Humans , Mice , Ovarian Neoplasms/pathology , Precancerous Conditions/pathology , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/genetics
9.
J Clin Invest ; 127(10): 3796-3809, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28891811

ABSTRACT

Macrophages are attracted to developing tumors and can participate in immune surveillance to eliminate neoplastic cells. In response, neoplastic cells utilize NF-κB to suppress this killing activity, but the mechanisms underlying their self-protection remain unclear. Here, we report that this dynamic interaction between tumor cells and macrophages is integrally linked by a soluble factor identified as growth and differentiation factor 15 (GDF-15). In vitro, tumor-derived GDF-15 signals in macrophages to suppress their proapoptotic activity by inhibiting TNF and nitric oxide (NO) production. In vivo, depletion of GDF-15 in Ras-driven tumor xenografts and in an orthotopic model of pancreatic cancer delayed tumor development. This delay correlated with increased infiltrating antitumor macrophages. Further, production of GDF-15 is directly regulated by NF-κB, and the colocalization of activated NF-κB and GDF-15 in epithelial ducts of human pancreatic adenocarcinoma supports the importance of this observation. Mechanistically, we found that GDF-15 suppresses macrophage activity by inhibiting TGF-ß-activated kinase (TAK1) signaling to NF-κB, thereby blocking synthesis of TNF and NO. Based on these results, we propose that the NF-κB/GDF-15 regulatory axis is important for tumor cells in evading macrophage immune surveillance during the early stages of tumorigenesis.


Subject(s)
Adenocarcinoma/immunology , Growth Differentiation Factor 15/immunology , Immunologic Surveillance , Macrophages/immunology , NF-kappa B/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Pancreatic Neoplasms/immunology , Signal Transduction/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Female , Growth Differentiation Factor 15/genetics , Heterografts , MAP Kinase Kinase Kinases , Macrophages/pathology , Male , Mice , Mice, Knockout , NF-kappa B/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Nitric Oxide/genetics , Nitric Oxide/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
J Immunol ; 199(7): 2333-2342, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28842466

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are important regulators of the immune system, maintaining homeostasis in the presence of commensal bacteria, but activating immune defenses in response to microbial pathogens. ILC3s are a robust source of IL-22, a cytokine critical for stimulating the antimicrobial response. We sought to identify cytokines that can promote proliferation and induce or maintain IL-22 production by ILC3s and determine a molecular mechanism for this process. We identified IL-18 as a cytokine that cooperates with an ILC3 survival factor, IL-15, to induce proliferation of human ILC3s, as well as induce and maintain IL-22 production. To determine a mechanism of action, we examined the NF-κB pathway, which is activated by IL-18 signaling. We found that the NF-κB complex signaling component, p65, binds to the proximal region of the IL22 promoter and promotes transcriptional activity. Finally, we observed that CD11c+ dendritic cells expressing IL-18 are found in close proximity to ILC3s in human tonsils in situ. Therefore, we identify a new mechanism by which human ILC3s proliferate and produce IL-22, and identify NF-κB as a potential therapeutic target to be considered in pathologic states characterized by overproduction of IL-18 and/or IL-22.


Subject(s)
Cell Proliferation , Interleukin-18/metabolism , Interleukins/biosynthesis , Lymphocytes/physiology , NF-kappa B/metabolism , Signal Transduction , Dendritic Cells/physiology , Humans , Immunity, Innate , Interleukin-15/immunology , Interleukins/genetics , Interleukins/immunology , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Promoter Regions, Genetic , Signal Transduction/immunology , Transcription Factor RelA/metabolism , Interleukin-22
11.
Cancer Res ; 77(14): 3846-3856, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28588009

ABSTRACT

Despite the development of combined modality treatments against liposarcoma in recent years, a significant proportion of patients respond only modestly to such approaches, possibly contributing to local or distant recurrence. Early detection of recurrent or metastatic disease could improve patient prognosis by triggering earlier clinical intervention. However, useful biomarkers for such purposes are lacking. Using both patient plasma samples and cell lines, we demonstrate here that miR-25-3p and miR-92a-3p are secreted by liposarcoma cells through extracellular vesicles and may be useful as potential biomarkers of disease. Both miR-25-3p and miR-92a-3p stimulated secretion of proinflammatory cytokine IL6 from tumor-associated macrophages in a TLR7/8-dependent manner, which in turn promoted liposarcoma cell proliferation, invasion, and metastasis via this interaction with the surrounding microenvironment. Our findings provide novel and previously unreported insight into liposarcoma progression, identifying communication between liposarcoma cells and their microenvironment as a process critically involved in liposarcoma progression. This study establishes the possibility that the pattern of circulating miRNAs may identify recurrence prior to radiological detectability while providing insight into disease outcome and as a possible approach to monitor treatment efficacy. Cancer Res; 77(14); 3846-56. ©2017 AACR.


Subject(s)
Exosomes/metabolism , Liposarcoma/genetics , MicroRNAs/metabolism , Animals , Case-Control Studies , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , HEK293 Cells , Humans , Liposarcoma/blood , Liposarcoma/pathology , Mice , Mice, Inbred C57BL , Transfection
12.
Neoplasia ; 19(6): 496-508, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28501760

ABSTRACT

The extracellular matrix (ECM) is critical for mammary ductal development and differentiation, but how mammary fibroblasts regulate ECM remodeling remains to be elucidated. Herein, we used a mouse genetic model to activate platelet derived growth factor receptor-alpha (PDGFRα) specifically in the stroma. Hyperactivation of PDGFRα in the mammary stroma severely hindered pubertal mammary ductal morphogenesis, but did not interrupt the lobuloalveolar differentiation program. Increased stromal PDGFRα signaling induced mammary fat pad fibrosis with a corresponding increase in interstitial hyaluronic acid (HA) and collagen deposition. Mammary fibroblasts with PDGFRα hyperactivation also decreased hydraulic permeability of a collagen substrate in an in vitro microfluidic device assay, which was mitigated by inhibition of either PDGFRα or HA. Fibrosis seen in this model significantly increased the overall stiffness of the mammary gland as measured by atomic force microscopy. Further, mammary tumor cells injected orthotopically in the fat pads of mice with stromal activation of PDGFRα grew larger tumors compared to controls. Taken together, our data establish that aberrant stromal PDGFRα signaling disrupts ECM homeostasis during mammary gland development, resulting in increased mammary stiffness and increased potential for tumor growth.


Subject(s)
Mammary Glands, Animal/growth & development , Mammary Glands, Human/growth & development , Mammary Neoplasms, Animal/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Animals , Cell Differentiation/genetics , Extracellular Matrix/genetics , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Hyaluronic Acid/administration & dosage , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Mammary Neoplasms, Animal/pathology , Mice , Morphogenesis/genetics , Signal Transduction , Stromal Cells/pathology
13.
Neoplasia ; 18(9): 541-52, 2016 09.
Article in English | MEDLINE | ID: mdl-27659014

ABSTRACT

Preclinical studies have suggested that the pancreatic tumor microenvironment both inhibits and promotes tumor development and growth. Here we establish the role of stromal fibroblasts during acinar-to-ductal metaplasia (ADM), an initiating event in pancreatic cancer formation. The transcription factor V-Ets avian erythroblastosis virus E26 oncogene homolog 2 (ETS2) was elevated in smooth muscle actin-positive fibroblasts in the stroma of pancreatic ductal adenocarcinoma (PDAC) patient tissue samples relative to normal pancreatic controls. LSL-Kras(G12D/+); LSL-Trp53(R172H/+); Pdx-1-Cre (KPC) mice showed that ETS2 expression initially increased in fibroblasts during ADM and remained elevated through progression to PDAC. Conditional ablation of Ets-2 in pancreatic fibroblasts in a Kras(G12D)-driven mouse ADM model decreased the amount of ADM events. ADMs from fibroblast Ets-2-deleted animals had reduced epithelial cell proliferation and increased apoptosis. Surprisingly, fibroblast Ets-2 deletion significantly altered immune cell infiltration into the stroma, with an increased CD8+ T-cell population, and decreased presence of regulatory T cells (Tregs), myeloid-derived suppressor cells, and mature macrophages. The mechanism involved ETS2-dependent chemokine ligand production in fibroblasts. ETS2 directly bound to regulatory sequences for Ccl3, Ccl4, Cxcl4, Cxcl5, and Cxcl10, a group of chemokines that act as potent mediators of immune cell recruitment. These results suggest an unappreciated role for ETS2 in fibroblasts in establishing an immune-suppressive microenvironment in response to oncogenic Kras(G12D) signaling during the initial stages of tumor development.


Subject(s)
Acinar Cells/metabolism , Cell Transformation, Neoplastic/metabolism , Chemokines/biosynthesis , Chemotaxis, Leukocyte , Pancreatic Ducts/metabolism , Proto-Oncogene Protein c-ets-2/metabolism , Stromal Cells/metabolism , Acinar Cells/pathology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Chemotaxis, Leukocyte/immunology , Collagen/metabolism , Fibroblasts/metabolism , Gene Deletion , Gene Expression , Humans , Immunohistochemistry , Metaplasia , Mice , Mice, Knockout , Pancreatic Ducts/immunology , Pancreatic Ducts/pathology , Phenotype , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction
14.
Genes Dev ; 30(17): 1943-55, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27633013

ABSTRACT

The contribution of the microenvironment to pancreatic acinar-to-ductal metaplasia (ADM), a preneoplastic transition in oncogenic Kras-driven pancreatic cancer progression, is currently unclear. Here we show that disruption of paracrine Hedgehog signaling via genetic ablation of Smoothened (Smo) in stromal fibroblasts in a Kras(G12D) mouse model increased ADM. Smo-deleted fibroblasts had higher expression of transforming growth factor-α (Tgfa) mRNA and secreted higher levels of TGFα, leading to activation of EGFR signaling in acinar cells and increased ADM. The mechanism involved activation of AKT and noncanonical activation of the GLI family transcription factor GLI2. GLI2 was phosphorylated at Ser230 in an AKT-dependent fashion and directly regulated Tgfa expression in fibroblasts lacking Smo Additionally, Smo-deleted fibroblasts stimulated the growth of Kras(G12D)/Tp53(R172H) pancreatic tumor cells in vivo and in vitro. These results define a non-cell-autonomous mechanism modulating Kras(G12D)-driven ADM that is balanced by cross-talk between Hedgehog/SMO and AKT/GLI2 pathways in stromal fibroblasts.


Subject(s)
Carcinoma, Pancreatic Ductal , Metaplasia/genetics , Metaplasia/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/pathology , Gene Deletion , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Pancreas/pathology , Signal Transduction/genetics , Transforming Growth Factor alpha/metabolism , Tumor Cells, Cultured , Zinc Finger Protein Gli2
15.
Genes Dev ; 29(16): 1707-20, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26302789

ABSTRACT

Inactivation of phosphatase and tensin homology deleted on chromosome 10 (PTEN) is linked to increased PI3K-AKT signaling, enhanced organismal growth, and cancer development. Here we generated and analyzed Pten knock-in mice harboring a C2 domain missense mutation at phenylalanine 341 (Pten(FV)), found in human cancer. Despite having reduced levels of PTEN protein, homozygous Pten(FV/FV) embryos have intact AKT signaling, develop normally, and are carried to term. Heterozygous Pten(FV/+) mice develop carcinoma in the thymus, stomach, adrenal medulla, and mammary gland but not in other organs typically sensitive to Pten deficiency, including the thyroid, prostate, and uterus. Progression to carcinoma in sensitive organs ensues in the absence of overt AKT activation. Carcinoma in the uterus, a cancer-resistant organ, requires a second clonal event associated with the spontaneous activation of AKT and downstream signaling. In summary, this PTEN noncatalytic missense mutation exposes a core tumor suppressor function distinct from inhibition of canonical AKT signaling that predisposes to organ-selective cancer development in vivo.


Subject(s)
Carcinoma/genetics , Mutation, Missense/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , Carcinoma/enzymology , Carcinoma/physiopathology , Cell Nucleus/metabolism , Cells, Cultured , Embryo, Mammalian , Enzyme Activation , Female , Gene Knock-In Techniques , Mice , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Protein Stability
16.
PLoS One ; 8(8): e71533, 2013.
Article in English | MEDLINE | ID: mdl-23977064

ABSTRACT

Tumor fibroblasts are active partners in tumor progression, but the genes and pathways that mediate this collaboration are ill-defined. Previous work demonstrates that Ets2 function in stromal cells significantly contributes to breast tumor progression. Conditional mouse models were used to study the function of Ets2 in both mammary stromal fibroblasts and epithelial cells. Conditional inactivation of Ets2 in stromal fibroblasts in PyMT and ErbB2 driven tumors significantly reduced tumor growth, however deletion of Ets2 in epithelial cells in the PyMT model had no significant effect. Analysis of gene expression in fibroblasts revealed a tumor- and Ets2-dependent gene signature that was enriched in genes important for ECM remodeling, cell migration, and angiogenesis in both PyMT and ErbB2 driven-tumors. Consistent with these results, PyMT and ErbB2 tumors lacking Ets2 in fibroblasts had fewer functional blood vessels, and Ets2 in fibroblasts elicited changes in gene expression in tumor endothelial cells consistent with this phenotype. An in vivo angiogenesis assay revealed the ability of Ets2 in fibroblasts to promote blood vessel formation in the absence of tumor cells. Importantly, the Ets2-dependent gene expression signatures from both mouse models were able to distinguish human breast tumor stroma from normal stroma, and correlated with patient outcomes in two whole tumor breast cancer data sets. The data reveals a key function for Ets2 in tumor fibroblasts in signaling to endothelial cells to promote tumor angiogenesis. The results highlight the collaborative networks that orchestrate communication between stromal cells and tumor cells, and suggest that targeting tumor fibroblasts may be an effective strategy for developing novel anti-angiogenic therapies.


Subject(s)
Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Proto-Oncogene Protein c-ets-2/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Compartmentation , Disease Models, Animal , Disease Progression , Female , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Stromal Cells/metabolism , Stromal Cells/pathology , Treatment Outcome
17.
PLoS One ; 7(7): e42005, 2012.
Article in English | MEDLINE | ID: mdl-22860046

ABSTRACT

Osteoclasts are terminally differentiated cells that attach to bone and secrete proteases to degrade the bone matrix. The primary protease responsible for the degradation of the organic component of the bone matrix is Cathepsin K, which was largely thought to be unique to osteoclasts. Given its apparent selective expression in osteoclasts, the Cathepsin K promoter has been engineered to drive the expression of Cre recombinase in mice and has been the most relevant tool for generating osteoclast-specific gene loss. In an effort to understand the role of the ARF tumor suppressor in osteoclasts, we crossed Arf (fl/fl) mice to Ctsk(Cre/+) mice, which unexpectedly resulted in the germline loss of Arf. We subsequently confirmed Cre activity in gametes by generating Ctsk(Cre/+); Rosa(+) mice. These results raise significant concerns regarding in vivo bone phenotypes created using Ctsk(Cre/+) mice and warrant further investigation into the role of Cathepsin K in gametes as well as alternative tools for studying osteoclast-specific gene loss in vivo.


Subject(s)
Cathepsin K/genetics , Gene Deletion , Germ Cells , Integrases/genetics , Animals , Base Sequence , DNA Primers , Female , Male , Mice , Real-Time Polymerase Chain Reaction
18.
Mol Cancer Res ; 9(12): 1686-95, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21970857

ABSTRACT

Trop2, an oncogenic cell surface protein under investigation as a therapeutic target, is commonly overexpressed in several epithelial tumor types yet its function in tumor biology remains relatively unexplored. To investigate the role of Trop2 in epithelial carcinogenesis, we generated Trop2(-/-) mice, which are viable and possess a normal lifespan. Contrary to expectations, Trop2 loss fails to suppress keratinocyte transformation. Instead, ras-transformed Trop2(-/-) keratinocytes preferentially pass through an epithelial to mesenchymal transition (EMT) and form tumors with spindle cell histology. Furthermore, Trop2 loss renders Arf-null mice susceptible to the formation of biphasic sarcomatoid carcinomas containing both squamous and spindle cell components upon carcinogen exposure in an otherwise skin cancer-resistant strain (C57BL/6). Immortalized keratinocytes derived from Trop2(-/-)Arf(-/-) mice exhibit enhanced proliferative and migratory capacity as well as increased activation of mitogen-activated protein kinase and Src prior to transformation. The clinical relevance of these findings was supported by studying the molecular epidemiology of Trop2 in primary head and neck squamous cell carcinomas. This analysis revealed that Trop2 mRNA levels are decreased in a subset of tumors with features of EMT, and total loss of Trop2 protein expression is observed in the spindle cell component of sarcomatoid carcinomas. Therefore, while previous studies have emphasized the potential importance of Trop2 gain of function, these results uncover a role for Trop2 loss in tumorigenesis and the mesenchymal transdifferentiation observed in a subset of squamous cell carcinomas.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , 9,10-Dimethyl-1,2-benzanthracene/chemistry , Animals , Carcinoma, Squamous Cell/genetics , Cell Dedifferentiation , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Female , Gene Expression Regulation, Neoplastic , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/chemistry
19.
Cancer Res ; 71(21): 6708-17, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21937682

ABSTRACT

The p19ARF tumor suppressor limits ribosome biogenesis and responds to hyperproliferative signals to activate the p53 checkpoint response. Although its activation of p53 has been well characterized, the role of ARF in restraining nucleolar ribosome production is poorly understood. Here we report the use of a mass spectroscopic analysis to identify protein changes within the nucleoli of Arf-deficient mouse cells. Through this approach, we discovered that ARF limited the nucleolar localization of the RNA helicase DDX5, which promotes the synthesis and maturation of rRNA, ultimately increasing ribosome output and proliferation. ARF inhibited the interaction between DDX5 and nucleophosmin (NPM), preventing association of DDX5 with the rDNA promoter and nuclear pre-ribosomes. In addition, Arf-deficient cells transformed by oncogenic RasV12 were addicted to DDX5, because reduction of DDX5 was sufficient to impair RasV12-driven colony formation in soft agar and tumor growth in mice. Taken together, our findings indicate that DDX5 is a key p53-independent target of the ARF tumor suppressor and is a novel non-oncogene participant in ribosome biogenesis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/physiology , DEAD-box RNA Helicases/physiology , Ribosomes/metabolism , Animals , Cell Division , Cell Nucleolus/metabolism , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/deficiency , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Fibroblasts , Mice , Mice, Knockout , Mice, Nude , Nuclear Proteins/metabolism , Nucleophosmin , Oncogene Protein p21(ras)/physiology , Promoter Regions, Genetic , Protein Transport , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/physiology
20.
Cancer Res ; 70(21): 8937-47, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20940396

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway regulates mammalian cell growth, survival, and motility and plays a major pathogenetic role in human prostate cancer (PCa). However, the oncogenic contributions downstream of the PI3K pathway made by mammalian target of rapamycin complex 1 (mTORC1)-mediated cell growth signal transduction in PCa have yet to be elucidated in detail. Here, we engineered constitutive mTORC1 activation in prostate epithelium by a conditional genetic deletion of tuberous sclerosis complex 1 (Tsc1), a potent negative regulator of mTORC1 signaling. Epithelial inactivation was not immediately tumorigenic, but Tsc1-deficient mice developed prostatic intraepithelial neoplasia (mPIN) in lateral and anterior prostates by 6 months of age, with increasing disease penetrance over time. Lateral prostate lesions in 16- to 22-month-old mutant mice progressed to two types of more advanced lesions, adenomatous gland forming lesion (Type 1) and atypical glands embedded in massively expanded reactive stroma (Type 2). Both Type 1 and Type 2 lesions contained multiple foci of microinvasive carcinoma. Epithelial neoplastic and atypical stromal lesions persisted despite 4 weeks of RAD001 chemotherapy. Rapalogue resistance was not due to AKT or extracellular signal-regulated kinase 1/2 activation. Expression of the homeobox gene Nkx3.1 was lost in Tsc1-deficient mPIN, and it cooperated with TSC1 loss in mPIN initiation in doubly mutant Tsc1:Nkx3.1 prostatic epithelial knockout mice. Thus, TSC1 inactivation distal to PI3K and AKT activation is sufficient to activate a molecular signaling cascade producing prostatic neoplasia and focal carcinogenesis.


Subject(s)
Genes, Tumor Suppressor , Homeodomain Proteins/physiology , Prostate/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Blotting, Western , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 3/metabolism , Multiprotein Complexes , Phosphatidylinositol 3-Kinase/metabolism , Prostate/metabolism , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Neoplasms/genetics , Proteins/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...