Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(13): eadf0927, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36989360

ABSTRACT

Cell state plasticity is carefully regulated in adult epithelia to prevent cancer. The aberrant expansion of the normally restricted capability for cell state plasticity in neoplasia is poorly defined. Using genetically engineered and carcinogen-induced mouse models of intestinal neoplasia, we observed that impaired differentiation is a conserved event preceding cancer development. Single-cell RNA sequencing (scRNA-seq) of premalignant lesions from mouse models and a patient with hereditary polyposis revealed that cancer initiates by adopting an aberrant transcriptional state characterized by regenerative activity, marked by Ly6a (Sca-1), and reactivation of fetal intestinal genes, including Tacstd2 (Trop2). Genetic inactivation of Sox9 prevented adenoma formation, obstructed the emergence of regenerative and fetal programs, and restored multilineage differentiation by scRNA-seq. Expanded chromatin accessibility at regeneration and fetal genes upon Apc inactivation was reduced by concomitant Sox9 suppression. These studies indicate that aberrant cell state plasticity mediated by unabated regenerative activity and developmental reprogramming precedes cancer development.


Subject(s)
Adenoma , Colorectal Neoplasms , Mice , Animals , Intestines , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cell Differentiation , Adenoma/genetics , Adenoma/pathology
2.
PLoS Genet ; 18(9): e1010362, 2022 09.
Article in English | MEDLINE | ID: mdl-36054194

ABSTRACT

The role of EGFR in lung cancer is well described with numerous activating mutations that result in phosphorylation and tyrosine kinase inhibitors that target EGFR. While the role of the EGFR kinase in non-small cell lung cancer (NSCLC) is appreciated, control of EGFR signaling pathways through dephosphorylation by phosphatases is not as clear. Through whole genome sequencing we have uncovered conserved V483M Ptprh mutations in PyMT induced tumors. Profiling the downstream events of Ptprh mutant tumors revealed AKT activation, suggesting a key target of PTPRH was EGFR tyrosine 1197. Given the role of EGFR in lung cancer, we explored TCGA data which revealed that a subset of PTPRH mutant tumors shared gene expression profiles with EGFR mutant tumors, but that EGFR mutations and PTPRH mutations were mutually exclusive. Generation of a PTPRH knockout NSCLC cell line resulted in Y1197 phosphorylation of EGFR, and a rescue with expression of wild type PTPRH returned EGFR phosphorylation to parental line values while rescue with catalytically dead PTPRH did not. A dose response curve illustrated that two human NSCLC lines with naturally occurring PTPRH mutations responded to EGFR tyrosine kinase inhibition. Osimertinib treatment of these tumors resulted in a reduction of tumor volume relative to vehicle controls. PTPRH mutation resulted in nuclear pEGFR as seen in immunohistochemistry, suggesting that there may also be a role for EGFR as a transcriptional co-factor. Together these data suggest mutations in PTPRH in NSCLC is inhibitory to PTPRH function, resulting in aberrant EGFR activity and ultimately may result in clinically actionable alterations using existing therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Tyrosine/genetics
3.
Elife ; 112022 05 12.
Article in English | MEDLINE | ID: mdl-35550030

ABSTRACT

Metastatic castration-resistant prostate cancers (mCRPCs) are treated with therapies that antagonize the androgen receptor (AR). Nearly all patients develop resistance to AR-targeted therapies (ARTs). Our previous work identified CREB5 as an upregulated target gene in human mCRPC that promoted resistance to all clinically approved ART. The mechanisms by which CREB5 promotes progression of mCRPC or other cancers remains elusive. Integrating ChIP-seq and rapid immunoprecipitation and mass spectroscopy of endogenous proteins, we report that cells overexpressing CREB5 demonstrate extensive reprogramming of nuclear protein-protein interactions in response to the ART agent enzalutamide. Specifically, CREB5 physically interacts with AR, the pioneering actor FOXA1, and other known co-factors of AR and FOXA1 at transcription regulatory elements recently found to be active in mCRPC patients. We identified a subset of CREB5/FOXA1 co-interacting nuclear factors that have critical functions for AR transcription (GRHL2, HOXB13) while others (TBX3, NFIC) regulated cell viability and ART resistance and were amplified or overexpressed in mCRPC. Upon examining the nuclear protein interactions and the impact of CREB5 expression on the mCRPC patient transcriptome, we found that CREB5 was associated with Wnt signaling and epithelial to mesenchymal transitions, implicating these pathways in CREB5/FOXA1-mediated ART resistance. Overall, these observations define the molecular interactions among CREB5, FOXA1, and pathways that promote ART resistance.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein A , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
4.
Dev Cell ; 57(2): 212-227.e8, 2022 01 24.
Article in English | MEDLINE | ID: mdl-34990589

ABSTRACT

The transcriptional co-activator YAP1 oncogene is the downstream effector of the Hippo pathway, which regulates tissue homeostasis, organ size, regeneration, and tumorigenesis. Multiple cancers are dependent on sustained expression of YAP1 for cell proliferation, survival, and tumorigenesis, but the molecular basis of this oncogene dependency is not well understood. To identify genes that can functionally substitute for YAP1, we performed a genome-scale genetic rescue screen in YAP1-dependent colon cancer cells expressing an inducible YAP1-specific shRNA. We found that the transcription factor PRDM14 rescued cell proliferation and tumorigenesis upon YAP1 suppression in YAP1-dependent cells, xenografts, and colon cancer organoids. YAP1 and PRDM14 individually activated the transcription of calmodulin 2 (CALM2) and a glucose transporter SLC2A1 upon YAP1 suppression, and CALM2 or SLC2A1 expression was required for the rescue of YAP1 suppression. Together, these findings implicate PRDM14-mediated transcriptional upregulation of CALM2 and SLC2A1 as key components of oncogenic YAP1 signaling and dependency.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calmodulin/genetics , Calmodulin/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Glucose Transporter Type 1/genetics , Humans , Mice , Mice, Nude , Organoids , Phosphoproteins/metabolism , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Transcriptional Activation , Xenograft Model Antitumor Assays , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/physiology
5.
Cell Rep ; 36(4): 109443, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34320363

ABSTRACT

Metastasis is a complex and poorly understood process. In pancreatic cancer, loss of the transforming growth factor (TGF)-ß/BMP effector SMAD4 is correlated with changes in altered histopathological transitions, metastatic disease, and poor prognosis. In this study, we use isogenic cancer cell lines to identify SMAD4 regulated genes that contribute to the development of metastatic colonization. We perform an in vivo screen identifying FOSL1 as both a SMAD4 target and sufficient to drive colonization to the lung. The targeting of these genes early in treatment may provide a therapeutic benefit.


Subject(s)
Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-fos/genetics , Smad4 Protein/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Enhancer Elements, Genetic/genetics , Humans , Mice , Neoplasm Metastasis , Proto-Oncogene Proteins c-fos/metabolism , Pancreatic Neoplasms
6.
Int J Mol Sci ; 21(22)2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33217970

ABSTRACT

Sarcomere and cytoskeleton genes, or actomyosin genes, regulate cell biology including mechanical stress, cell motility, and cell division. While actomyosin genes are recurrently dysregulated in cancers, their oncogenic roles have not been examined in a lineage-specific fashion. In this report, we investigated dysregulation of nine sarcomeric and cytoskeletal genes across 20 cancer lineages. We found that uterine cancers harbored the highest frequencies of amplification and overexpression of the gamma actin gene, ACTG1. Each of the four subtypes of uterine cancers, mixed endometrial carcinomas, serous carcinomas, endometroid carcinomas, and carcinosarcomas harbored between 5~20% of ACTG1 gene amplification or overexpression. Clinically, patients with ACTG1 gains had a poor prognosis. ACTG1 gains showed transcriptional patterns that reflect activation of oncogenic signals, repressed response to innate immunity, or immunotherapy. Functionally, the CRISPR-CAS9 gene deletion of ACTG1 had the most robust and consistent effects in uterine cancer cells relative to 20 other lineages. Overall, we propose that ACTG1 regulates the fitness of uterine cancer cells by modulating cell-intrinsic properties and the tumor microenvironment. In summary, the ACTG1 functions relative to other actomyosin genes support the notion that it is a potential biomarker and a target gene in uterine cancer precision therapies.


Subject(s)
Actins , Biomarkers, Tumor , Gene Amplification , Neoplasm Proteins , Uterine Neoplasms , Actins/genetics , Actins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Survival Rate , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Uterine Neoplasms/mortality , Uterine Neoplasms/pathology
7.
Sci Rep ; 10(1): 17948, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087787

ABSTRACT

The E2F family, classically known for a central role in cell cycle, has a number of emerging roles in cancer including angiogenesis, metabolic reprogramming, metastasis and DNA repair. E2F1 specifically has been shown to be a critical mediator of DNA repair; however, little is known about DNA repair and other E2F family members. Here we present an integrative bioinformatic and high throughput drug screening study to define the role of E2F2 in maintaining genomic integrity in breast cancer. We utilized in vitro E2F2 ChIP-chip and over expression data to identify transcriptional targets of E2F2. This data was integrated with gene expression from E2F2 knockout tumors in an MMTV-Neu background. Finally, this data was compared to human datasets to identify conserved roles of E2F2 in human breast cancer through the TCGA breast cancer, Cancer Cell Line Encyclopedia, and CancerRx datasets. Through these methods we predict that E2F2 transcriptionally regulates mediators of DNA repair. Our gene expression data supports this hypothesis and low E2F2 activity is associated with a highly unstable tumor. In human breast cancer E2F2, status was also correlated with a patient's response to PARP inhibition therapy. Taken together this manuscript defines a novel role of E2F2 in cancer progression beyond cell cycle and could impact patient treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , E2F2 Transcription Factor/physiology , Genomic Instability/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Animals , Breast Neoplasms/drug therapy , Cell Cycle/genetics , Cell Line, Tumor , DNA Repair/genetics , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Neovascularization, Pathologic/genetics
8.
Adv Physiol Educ ; 44(3): 459-463, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32795123

ABSTRACT

The laboratory course is an excellent venue to apply content, practice inquiry, improve critical thinking, practice key clinical skills, and work with data. The use of inquiry-based course projects allows for students to propose open ended questions, form a hypothesis, design an experiment, collect data, analyze data, draw conclusion, and present their findings. This comprehensive experience is ideal for a capstone (senior level) laboratory course that is the culmination of 4 yr of study in the degree. At Michigan State University, the capstone laboratory has incorporated a formal course-based research experience in human physiology. The rationale and logistics for running such an experience are described in this paper.


Subject(s)
Curriculum , Laboratories , Physiology , Humans , Physiology/education , Students , Thinking , Universities
9.
Cell Rep ; 29(3): 589-602.e6, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618629

ABSTRACT

Integrin receptors coordinate cell adhesion to the extracellular matrix (ECM) to facilitate many cellular processes during malignant transformation. Despite their pro-tumorigenic roles, therapies targeting integrins remain limited. Here, we provide genetic evidence supporting a functional redundancy between ß1 and ß3 integrin during breast cancer progression. Although ablation of ß1 or ß3 integrin alone has limited effects on ErbB2-driven mammary tumorigenesis, deletion of both receptors resulted in a significant delay in tumor onset with a corresponding impairment in lung metastasis. Mechanistically, stiff ECM cooperates with integrin receptors to recruit insulin receptors (IRs) to focal adhesion through the formation of integrin/IR complexes, thereby preventing their lysosomal degradation. ß1/ß3 integrin-deficient tumors that eventually emerged exhibit impaired Akt/mTORC1 activity. Murine and human breast cancers exhibiting enhanced integrin-dependent activity also display elevated IR/Akt/mTORC1 signaling activity. Together, these observations argue that integrin/IR crosstalk transduces mechanical cues from the tumor microenvironment to promote ErbB2-dependent breast cancer progression.


Subject(s)
Breast Neoplasms/pathology , Integrin beta1/metabolism , Integrin beta3/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Adult , Animals , Breast Neoplasms/mortality , Cell Adhesion , Extracellular Matrix/metabolism , Female , Humans , Insulin/pharmacology , Integrin beta1/genetics , Integrin beta3/genetics , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Tumor Microenvironment
10.
Cell Rep ; 29(2): 249-257.e8, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31597089

ABSTRACT

Monoclonal antibodies (mAbs) targeting the oncogenic receptor tyrosine kinase ERBB2/HER2, such as Trastuzumab, are the standard of care therapy for breast cancers driven by ERBB2 overexpression and activation. However, a substantial proportion of patients exhibit de novo resistance. Here, by comparing matched Trastuzumab-naive and post-treatment patient samples from a neoadjuvant trial, we link resistance with elevation of H3K27me3, a repressive histone modification catalyzed by polycomb repressor complex 2 (PRC2). In ErbB2+ breast cancer models, PRC2 silences endogenous retroviruses (ERVs) to suppress anti-tumor type-I interferon (IFN) responses. In patients, elevated H3K27me3 in tumor cells following Trastuzumab treatment correlates with suppression of interferon-driven viral defense gene expression signatures and poor response. Using an immunocompetent model, we provide evidence that EZH2 inhibitors promote interferon-driven immune responses that enhance the efficacy of anti-ErbB2 mAbs, suggesting the potential clinical benefit of epigenomic reprogramming by H3K27me3 depletion in Trastuzumab-resistant disease.


Subject(s)
Histones/metabolism , Lysine/metabolism , Molecular Targeted Therapy , Receptor, ErbB-2/metabolism , Adult , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Humans , Interferon Type I/metabolism , Methylation , Mice , Models, Biological , Polycomb Repressive Complex 2/metabolism , Retroelements/genetics , Trastuzumab/therapeutic use , Up-Regulation
11.
Cell Rep ; 29(1): 118-134.e8, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577942

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathway is a critical effector of oncogenic RAS signaling, and MAPK pathway inhibition may be an effective combination treatment strategy. We performed genome-scale loss-of-function CRISPR-Cas9 screens in the presence of a MEK1/2 inhibitor (MEKi) in KRAS-mutant pancreatic and lung cancer cell lines and identified genes that cooperate with MEK inhibition. While we observed heterogeneity in genetic modifiers of MEKi sensitivity across cell lines, several recurrent classes of synthetic lethal vulnerabilities emerged at the pathway level. Multiple members of receptor tyrosine kinase (RTK)-RAS-MAPK pathways scored as sensitizers to MEKi. In particular, we demonstrate that knockout, suppression, or degradation of SHOC2, a positive regulator of MAPK signaling, specifically cooperated with MEK inhibition to impair proliferation in RAS-driven cancer cells. The depletion of SHOC2 disrupted survival pathways triggered by feedback RTK signaling in response to MEK inhibition. Thus, these findings nominate SHOC2 as a potential target for combination therapy.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/metabolism , ras Proteins/metabolism , A549 Cells , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , HCT116 Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Hairless , Mice, SCID , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
12.
Nat Commun ; 10(1): 3261, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31332182

ABSTRACT

Mouse models have an essential role in cancer research, yet little is known about how various models resemble human cancer at a genomic level. Here, we complete whole genome sequencing and transcriptome profiling of two widely used mouse models of breast cancer, MMTV-Neu and MMTV-PyMT. Through integrative in vitro and in vivo studies, we identify copy number alterations in key extracellular matrix proteins including collagen 1 type 1 alpha 1 (COL1A1) and chondroadherin (CHAD) that drive metastasis in these mouse models. In addition to copy number alterations, we observe a propensity of the tumors to modulate tyrosine kinase-mediated signaling through mutation of phosphatases such as PTPRH in the MMTV-PyMT mouse model. Mutation in PTPRH leads to increased phospho-EGFR levels and decreased latency. These findings underscore the importance of understanding the complete genomic landscape of a mouse model and illustrate the utility this has in understanding human cancers.


Subject(s)
Breast Neoplasms/genetics , Disease Models, Animal , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/genetics , Animals , Cell Line, Tumor , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Extracellular Matrix Proteins/genetics , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mice , Mutation
13.
Nat Commun ; 10(1): 2901, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31263101

ABSTRACT

Dysregulation of histone modifications promotes carcinogenesis by altering transcription. Breast cancers frequently overexpress the histone methyltransferase EZH2, the catalytic subunit of Polycomb Repressor Complex 2 (PRC2). However, the role of EZH2 in this setting is unclear due to the context-dependent functions of PRC2 and the heterogeneity of breast cancer. Moreover, the mechanisms underlying PRC2 overexpression in cancer are obscure. Here, using multiple models of breast cancer driven by the oncogene ErbB2, we show that the tyrosine kinase c-Src links energy sufficiency with PRC2 overexpression via control of mRNA translation. By stimulating mitochondrial ATP production, c-Src suppresses energy stress, permitting sustained activation of the mammalian/mechanistic target of rapamycin complex 1 (mTORC1), which increases the translation of mRNAs encoding the PRC2 subunits Ezh2 and Suz12. We show that Ezh2 overexpression and activity are pivotal in ErbB2-mediated mammary tumourigenesis. These results reveal the hitherto unknown c-Src/mTORC1/PRC2 axis, which is essential for ErbB2-driven carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic , Polycomb Repressive Complex 2/genetics , Receptor, ErbB-2/metabolism , src-Family Kinases/metabolism , Adenosine Triphosphate/metabolism , Adult , Animals , Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase , Carcinogenesis , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Polycomb Repressive Complex 2/metabolism , Protein Biosynthesis , Receptor, ErbB-2/genetics , src-Family Kinases/genetics
14.
Sci Rep ; 9(1): 10718, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31341204

ABSTRACT

In prior work we demonstrated that loss of E2F transcription factors inhibits metastasis. Here we address the mechanisms for this phenotype and identify the E2F regulated genes that coordinate tumor cell metastasis. Transcriptomic profiling of E2F1 knockout tumors identified a role for E2F1 as a master regulator of a suite of pro-metastatic genes, but also uncovered E2F1 target genes with an unknown role in pulmonary metastasis. High expression of one of these genes, Fgf13, is associated with early human breast cancer metastasis in a clinical dataset. Together these data led to the hypothesis that Fgf13 is critical for breast cancer metastasis, and that upregulation of Fgf13 may partially explain how E2F1 promotes breast cancer metastasis. To test this hypothesis we ablated Fgf13 via CRISPR. Deletion of Fgf13 in a MMTV-PyMT breast cancer cell line reduces colonization of the lungs in a tail vein injection. In addition, loss of Fgf13 reduced in vitro cell migration, suggesting that Fgf13 may be critical for tumor cells to escape the primary tumor and to colonize the distal sites. The significance of this work is twofold: we have both uncovered genomic features by which E2F1 regulates metastasis and we have identified new pro-metastatic functions for the E2F1 target gene Fgf13.


Subject(s)
Cell Movement , E2F1 Transcription Factor/metabolism , Fibroblast Growth Factors/genetics , Mammary Neoplasms, Experimental/metabolism , Animals , Cell Line, Tumor , E2F1 Transcription Factor/genetics , Female , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Metastasis
15.
Proc Natl Acad Sci U S A ; 115(31): 7869-7878, 2018 07 31.
Article in English | MEDLINE | ID: mdl-29987050

ABSTRACT

Endocrine therapy resistance invariably develops in advanced estrogen receptor-positive (ER+) breast cancer, but the underlying mechanisms are largely unknown. We have identified C-terminal SRC kinase (CSK) as a critical node in a previously unappreciated negative feedback loop that limits the efficacy of current ER-targeted therapies. Estrogen directly drives CSK expression in ER+ breast cancer. At low CSK levels, as is the case in patients with ER+ breast cancer resistant to endocrine therapy and with the poorest outcomes, the p21 protein-activated kinase 2 (PAK2) becomes activated and drives estrogen-independent growth. PAK2 overexpression is also associated with endocrine therapy resistance and worse clinical outcome, and the combination of a PAK2 inhibitor with an ER antagonist synergistically suppressed breast tumor growth. Clinical approaches to endocrine therapy-resistant breast cancer must overcome the loss of this estrogen-induced negative feedback loop that normally constrains the growth of ER+ tumors.


Subject(s)
Breast Neoplasms/drug therapy , Estrogens/pharmacology , Neoplasm Proteins/biosynthesis , Receptors, Estrogen/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Receptors, Estrogen/genetics , p21-Activated Kinases/biosynthesis , p21-Activated Kinases/genetics , src-Family Kinases/biosynthesis , src-Family Kinases/genetics
16.
Antioxid Redox Signal ; 29(16): 1535-1552, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29634345

ABSTRACT

AIMS: Activation of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway in normal cells inhibits carcinogenesis, whereas constitutive activation of Nrf2 in cancer cells promotes tumor growth and chemoresistance. However, the effects of Nrf2 activation in immune cells during lung carcinogenesis are poorly defined and could either promote or inhibit cancer growth. Our studies were designed to evaluate tumor burden and identify immune cell populations in the lungs of Nrf2 knockout (KO) versus wild-type (WT) mice challenged with vinyl carbamate. RESULTS: Nrf2 KO mice developed lung tumors earlier than the WT mice and exhibited more and larger tumors over time, even at late stages. T cell populations were lower in the lungs of Nrf2 KO mice, whereas tumor-promoting macrophages and myeloid-derived suppressor cells were elevated in the lungs and spleen, respectively, of Nrf2 KO mice relative to WT mice. Moreover, 34 immune response genes were significantly upregulated in tumors from Nrf2 KO mice, especially a series of cytokines (Cxcl1, Csf1, Ccl9, Cxcl12, etc.) and major histocompatibility complex antigens that promote tumor growth. INNOVATION: Our studies discovered a novel immune signature, characterized by the infiltration of tumor-promoting immune cells, elevated cytokines, and increased expression of immune response genes in the lungs and tumors of Nrf2 KO mice. A complementary profile was also found in lung cancer patients, supporting the clinical significance of our findings. CONCLUSION: Overall, our results confirmed a protective role for Nrf2 in late-stage carcinogenesis and, unexpectedly, suggest that activation of Nrf2 in immune cells may be advantageous for preventing or treating lung cancer. Antioxid. Redox Signal.


Subject(s)
Lung Neoplasms/immunology , NF-E2-Related Factor 2/immunology , Signal Transduction/immunology , Animals , Female , Lung Neoplasms/pathology , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
17.
Breast Cancer Res ; 20(1): 9, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382358

ABSTRACT

BACKGROUND: The Fos-related antigen 1 (FRA-1) transcription factor promotes tumor cell growth, invasion and metastasis. Phosphorylation of FRA-1 increases protein stability and function. We identify a novel signaling axis that leads to increased phosphorylation of FRA-1, increased extracellular matrix (ECM)-induced breast cancer cell invasion and is prognostic of poor outcome in patients with breast cancer. METHODS: While characterizing five breast cancer cell lines derived from primary human breast tumors, we identified BRC-31 as a novel basal-like cell model that expresses elevated FRA-1 levels. We interrogated the functional contribution of FRA-1 and an upstream signaling axis in breast cancer cell invasion. We extended this analysis to determine the prognostic significance of this signaling axis in samples derived from patients with breast cancer. RESULTS: BRC-31 cells display elevated focal adhesion kinase (FAK), SRC and extracellular signal-regulated (ERK2) phosphorylation relative to luminal breast cancer models. Inhibition of this signaling axis, with pharmacological inhibitors, reduces the phosphorylation and stabilization of FRA-1. Elevated integrin αVß3 and uPAR expression in these cells suggested that integrin receptors might activate this FAK-SRC-ERK2 signaling. Transient knockdown of urokinase/plasminogen activator urokinase receptor (uPAR) in basal-like breast cancer cells grown on vitronectin reduces FRA-1 phosphorylation and stabilization; and uPAR and FRA-1 are required for vitronectin-induced cell invasion. In clinical samples, a molecular component signature consisting of vitronectin-uPAR-uPA-FRA-1 predicts poor overall survival in patients with breast cancer and correlates with an FRA-1 transcriptional signature. CONCLUSIONS: We have identified a novel signaling axis that leads to phosphorylation and enhanced activity of FRA-1, a transcription factor that is emerging as an important modulator of breast cancer progression and metastasis.


Subject(s)
Breast Neoplasms/genetics , Proto-Oncogene Proteins c-fos/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/genetics , Breast Neoplasms/pathology , Extracellular Matrix/genetics , Female , Humans , Integrin alphaVbeta3/administration & dosage , Integrin alphaVbeta3/genetics , MCF-7 Cells , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Signal Transduction/drug effects , Vitronectin/administration & dosage
18.
J Mammary Gland Biol Neoplasia ; 22(1): 71-84, 2017 03.
Article in English | MEDLINE | ID: mdl-28124185

ABSTRACT

Breast tumor heterogeneity has been well documented through the use of multiplatform -omic studies in human tumors. However, there is no integrative database to capture the heterogeneity within mouse models of breast cancer. This project identifies genomic copy number alterations (CNAs) in 600 tumors across 27 major mouse models of breast cancer through the application of a predictive algorithm to publicly available gene expression data. It was found that despite the presence of strong oncogenic drivers in most mouse models, CNAs are extremely common but heterogeneous both between models and within models. Many mouse CNA events are largely conserved in human tumors and in the mouse we show that they are associated with secondary tumor characteristics such as tumor histology, metastasis, as well as enhanced oncogenic signaling. These data serve as an important resource in guiding investigators when choosing a mouse model to understand the gene copy number changes relevant to human breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA Copy Number Variations/genetics , Gene Dosage/genetics , Mammary Neoplasms, Animal/genetics , Animals , Disease Models, Animal , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Genomics/methods , Humans , Mice
19.
Cancer Immunol Res ; 4(7): 600-10, 2016 07.
Article in English | MEDLINE | ID: mdl-27197066

ABSTRACT

The abundance and functional orientation of tumor-infiltrating lymphocytes in breast cancer is associated with distant metastasis-free survival, yet how this association is influenced by tumor phenotypic heterogeneity is poorly understood. Here, a bioinformatics approach defined tumor biologic attributes that influence this association and delineated tumor subtypes that may differ in their ability to sustain durable antitumor immune responses. A large database of breast tumor expression profiles and associated clinical data was compiled, from which the ability of phenotypic markers to significantly influence the prognostic performance of a classification model that incorporates immune cell-specific gene signatures was ascertained. Markers of cell proliferation and intrinsic molecular subtype reproducibly distinguished two breast cancer subtypes that we refer to as immune benefit-enabled (IBE) and immune benefit-disabled (IBD). The IBE tumors, comprised mostly of highly proliferative tumors of the basal-like, HER2-enriched, and luminal B subtypes, could be stratified by the immune classifier into significantly different prognostic groups, while IBD tumors could not, indicating the potential for productive engagement of metastasis-protective immunity in IBE tumors, but not in IBD tumors. The prognostic stratification in IBE was independent of conventional variables. Gene network analysis predicted the activation of TNFα/IFNγ signaling pathways in IBE tumors and the activation of the transforming growth factor-ß pathway in IBD tumors. This prediction supports a model in which breast tumors can be distinguished on the basis of their potential for metastasis-protective immune responsiveness. Whether IBE and IBD represent clinically relevant contexts for evaluating sensitivity to immunotherapeutic agents warrants further investigation. Cancer Immunol Res; 4(7); 600-10. ©2016 AACR.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunomodulation , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Neoplasm Metastasis , Neoplasm Staging , Phenotype , Prognosis , Proportional Hazards Models
20.
Mol Cell Biol ; 36(10): 1509-25, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26976638

ABSTRACT

ShcA is an important mediator of ErbB2- and transforming growth factor ß (TGF-ß)-induced breast cancer cell migration, invasion, and metastasis. We show that in the context of reduced ShcA levels, the bone morphogenetic protein (BMP) antagonist chordin-like 1 (Chrdl1) is upregulated in numerous breast cancer cells following TGF-ß stimulation. BMPs have emerged as important modulators of breast cancer aggressiveness, and we have investigated the ability of Chrdl1 to block BMP-induced increases in breast cancer cell migration and invasion. Breast cancer-derived conditioned medium containing elevated concentrations of endogenous Chrdl1, as well as medium containing recombinant Chrdl1, suppresses BMP4-induced signaling in multiple breast cancer cell lines. Live-cell migration assays reveal that BMP4 induces breast cancer migration, which is effectively blocked by Chrdl1. We demonstrate that BMP4 also stimulated breast cancer cell invasion and matrix degradation, in part, through enhanced metalloproteinase 2 (MMP2) and MMP9 activity that is antagonized by Chrdl1. Finally, high Chrdl1 expression was associated with better clinical outcomes in patients with breast cancer. Together, our data reveal that Chrdl1 acts as a negative regulator of malignant breast cancer phenotypes through inhibition of BMP signaling.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Breast Neoplasms/pathology , Culture Media, Conditioned/pharmacology , Eye Proteins/metabolism , Nerve Tissue Proteins/metabolism , Bone Morphogenetic Protein 4/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Eye Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Nerve Tissue Proteins/genetics , Prognosis , Transforming Growth Factor beta/pharmacology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...