Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Cell Rep ; 35(9): 109156, 2021 06 01.
Article En | MEDLINE | ID: mdl-34077726

RBM39 is a known splicing factor and coactivator. Here, we report that RBM39 functions as a master transcriptional regulator that interacts with the MLL1 complex to facilitate chromatin binding and H3K4 trimethylation in breast cancer cells. We identify RBM39 functional domains required for DNA and complex binding and show that the loss of RBM39 has widespread effects on H3K4me3 and gene expression, including key oncogenic pathways. RBM39's RNA recognition motif 3 (RRM3) functions as a dominant-negative domain; namely, it disrupts the complex and H3K4me trimethylation and expression of RBM/MLL1 target genes. RRM3-derived cell-penetrating peptides phenocopy the effects of the loss of RBM39 to decrease growth and survival of all major subtypes of breast cancer and yet are nontoxic to normal cells. These findings establish RBM39/MLL1 as a major contributor to the abnormal epigenetic landscape in breast cancer and lay the foundation for peptide-mediated cancer-specific therapy based on disruption of RBM39 epigenomic functions.


Breast Neoplasms/genetics , Breast Neoplasms/pathology , Epigenomics , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Peptides/metabolism , RNA-Binding Proteins/genetics , Transcription, Genetic , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/genetics , Cell Survival , Cell-Penetrating Peptides/metabolism , Female , HEK293 Cells , Histone Code , Histone-Lysine N-Methyltransferase/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice, Inbred NOD , Mice, SCID , Myeloid-Lymphoid Leukemia Protein/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Domains , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
2.
Nat Med ; 22(11): 1321-1329, 2016 11.
Article En | MEDLINE | ID: mdl-27775705

Triple-negative breast cancer (TNBC), in which cells lack expression of the estrogen receptor (ER), the progesterone receptor (PR) and the ERBB2 (also known as HER2) receptor, is the breast cancer subtype with the poorest outcome. No targeted therapy is available against this subtype of cancer owing to a lack of validated molecular targets. We previously reported that signaling involving MYC-an essential, pleiotropic transcription factor that regulates the expression of hundreds of genes-is disproportionally higher in triple-negative (TN) tumors than in receptor-positive (RP) tumors. Direct inhibition of the oncogenic transcriptional activity of MYC has been challenging to achieve. Here, by conducting a shRNA screen targeting the kinome, we identified PIM1, a non-essential serine-threonine kinase, in a synthetic lethal interaction with MYC. PIM1 expression was higher in TN tumors than in RP tumors and was associated with poor prognosis in patients with hormone- and HER2-negative tumors. Small-molecule PIM kinase inhibitors halted the growth of human TN tumors with elevated MYC expression in patient-derived tumor xenograft (PDX) and MYC-driven transgenic mouse models of breast cancer by inhibiting the oncogenic transcriptional activity of MYC and restoring the function of the endogenous cell cycle inhibitor, p27. Our findings warrant clinical evaluation of PIM kinase inhibitors in patients with TN tumors that have elevated MYC expression.


Carcinoma, Ductal, Breast/metabolism , Mammary Neoplasms, Experimental/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Humans , In Situ Nick-End Labeling , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mice, Transgenic , Microscopy, Fluorescence , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays
3.
Nat Med ; 22(4): 427-32, 2016 04.
Article En | MEDLINE | ID: mdl-26950360

Expression of the oncogenic transcription factor MYC is disproportionately elevated in triple-negative breast cancer (TNBC), as compared to estrogen receptor-, progesterone receptor- or human epidermal growth factor 2 receptor-positive (RP) breast cancer. We and others have shown that MYC alters metabolism during tumorigenesis. However, the role of MYC in TNBC metabolism remains mostly unexplored. We hypothesized that MYC-dependent metabolic dysregulation is essential for the growth of MYC-overexpressing TNBC cells and may identify new therapeutic targets for this clinically challenging subset of breast cancer. Using a targeted metabolomics approach, we identified fatty acid oxidation (FAO) intermediates as being dramatically upregulated in a MYC-driven model of TNBC. We also identified a lipid metabolism gene signature in patients with TNBC that were identified from The Cancer Genome Atlas database and from multiple other clinical data sets, implicating FAO as a dysregulated pathway that is critical for TNBC cell metabolism. We found that pharmacologic inhibition of FAO catastrophically decreased energy metabolism in MYC-overexpressing TNBC cells and blocked tumor growth in a MYC-driven transgenic TNBC model and in a MYC-overexpressing TNBC patient-derived xenograft. These findings demonstrate that MYC-overexpressing TNBC shows an increased bioenergetic reliance on FAO and identify the inhibition of FAO as a potential therapeutic strategy for this subset of breast cancer.


Carcinogenesis/genetics , Energy Metabolism/genetics , Fatty Acids/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Triple Negative Breast Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/genetics , Energy Metabolism/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism/genetics , Oxidation-Reduction , Proto-Oncogene Proteins c-myc/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Nature ; 526(7571): 131-5, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26416748

Despite major advances in understanding the molecular and genetic basis of cancer, metastasis remains the cause of >90% of cancer-related mortality. Understanding metastasis initiation and progression is critical to developing new therapeutic strategies to treat and prevent metastatic disease. Prevailing theories hypothesize that metastases are seeded by rare tumour cells with unique properties, which may function like stem cells in their ability to initiate and propagate metastatic tumours. However, the identity of metastasis-initiating cells in human breast cancer remains elusive, and whether metastases are hierarchically organized is unknown. Here we show at the single-cell level that early stage metastatic cells possess a distinct stem-like gene expression signature. To identify and isolate metastatic cells from patient-derived xenograft models of human breast cancer, we developed a highly sensitive fluorescence-activated cell sorting (FACS)-based assay, which allowed us to enumerate metastatic cells in mouse peripheral tissues. We compared gene signatures in metastatic cells from tissues with low versus high metastatic burden. Metastatic cells from low-burden tissues were distinct owing to their increased expression of stem cell, epithelial-to-mesenchymal transition, pro-survival, and dormancy-associated genes. By contrast, metastatic cells from high-burden tissues were similar to primary tumour cells, which were more heterogeneous and expressed higher levels of luminal differentiation genes. Transplantation of stem-like metastatic cells from low-burden tissues showed that they have considerable tumour-initiating capacity, and can differentiate to produce luminal-like cancer cells. Progression to high metastatic burden was associated with increased proliferation and MYC expression, which could be attenuated by treatment with cyclin-dependent kinase (CDK) inhibitors. These findings support a hierarchical model for metastasis, in which metastases are initiated by stem-like cells that proliferate and differentiate to produce advanced metastatic disease.


Breast Neoplasms/pathology , Disease Progression , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/pathology , Single-Cell Analysis , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Separation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Flow Cytometry , Gene Expression Profiling , Genes, myc/genetics , Humans , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Xenograft Model Antitumor Assays
5.
Oncoimmunology ; 2(9): e25670, 2013 Sep 01.
Article En | MEDLINE | ID: mdl-24327933

The MSP/RON signaling pathway favors the conversion of micrometastatic lesions to overt metastases by suppressing antitumor immune responses. The loss of RON functions in the host potentiates tumor-specific CD8+ T-cell responses, hence inhibiting the outgrowth of metastatic cancer cells. Thus, RON inhibitors may potentially prevent the outgrowth of micrometastases in cancer patients.

6.
Cancer Discov ; 3(7): 751-60, 2013 Jul.
Article En | MEDLINE | ID: mdl-23612011

Many "nonmetastatic" cancers have spawned undetectable metastases before diagnosis. Eventual outgrowth of these microscopic lesions causes metastatic relapse and death, yet the events that dictate when and how micrometastases convert to overt metastases are largely unknown. We report that macrophage-stimulating protein and its receptor, Ron, are key mediators in conversion of micrometastases to bona fide metastatic lesions through immune suppression. Genetic deletion of Ron tyrosine kinase activity specifically in the host profoundly blocked metastasis. Our data show that loss of Ron function promotes an effective antitumor CD8(+) T-cell response, which specifically inhibits outgrowth of seeded metastatic colonies. Treatment of mice with a Ron-selective kinase inhibitor prevented outgrowth of lung metastasis, even when administered after micrometastatic colonies had already been established. Our findings indicate that Ron inhibitors may hold potential to specifically prevent outgrowth of micrometastases in patients with cancer in the adjuvant setting.


Immunity/genetics , Neoplasm Micrometastasis/genetics , Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Micrometastasis/pathology , Neoplasms/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction
7.
Genes Cancer ; 2(7): 753-62, 2011 Jul.
Article En | MEDLINE | ID: mdl-22207901

Receptor tyrosine kinases (RTKs) have been the subject of intense investigation due to their widespread deregulation in cancer and the prospect of developing targeted therapeutics against these proteins. The Ron RTK has been implicated in tumor aggressiveness and is a developing target for therapy, but its function in tumor progression and metastasis is not fully understood. We examined Ron activity in human breast cancers and found striking predominance of an activated Ron isoform known as short-form Ron (sfRon), whose function in breast tumors has not been explored. We found that sfRon plays a significant role in aggressiveness of breast cancer in vitro and in vivo. sfRon expression was sufficient to convert slow-growing, nonmetastatic tumors into rapidly growing tumors that spontaneously metastasized to liver and bones. Mechanistic studies revealed that sfRon promotes epithelial-mesenchymal transition, invasion, tumor growth, and metastasis through interaction with p85, the regulatory subunit of phosphoinositide 3-kinase (PI3K). Inhibition of PI3K activity, or introduction of a single mutation in the p85 docking site on sfRon, completely eliminated the ability of sfRon to promote tumor growth, invasion, and metastasis. These findings reveal sfRon as an important new player in breast cancer and validate Ron and PI3K as therapeutic targets in this disease.

8.
Curr Drug Targets ; 11(9): 1157-68, 2010 Sep.
Article En | MEDLINE | ID: mdl-20545605

Macrophage Stimulating Protein (MSP) is the only known ligand for the receptor tyrosine kinase Ron. The MSP/Ron pathway is involved in several important biological processes, including macrophage activity, wound healing, and epithelial cell behavior. A role for MSP/Ron in breast cancer has recently been elucidated, wherein this pathway regulates tumor growth, angiogenesis, and metastasis. Here, we review the recent literature surrounding MSP/Ron function in tumor cells, inflammatory cells, and osteoclasts - cell types that often coexist in breast tumor microenvironments. We discuss the potential implications of MSP/Ron activity occurring concurrently in these cell types on tumor progression and metastasis. Lastly, we outline the potential for targeting MSP/Ron as a novel therapy for breast cancer, and for other cancer types.


Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Hepatocyte Growth Factor/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Breast Neoplasms/physiopathology , Cell Line, Tumor , Disease Progression , Epithelial Cells/metabolism , Female , Humans , Inflammation/physiopathology , Macrophages/physiology , Mice , Mice, Transgenic , Molecular Targeted Therapy , Neoplasms/drug therapy , Osteoclasts/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Wound Healing
...