Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Breast Cancer Res ; 26(1): 56, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553763

ABSTRACT

Patient-derived organoid models of estrogen receptor-positive (ER+) breast cancer would provide a much-needed tool to understand drug resistance and disease progression better. However, the establishment and long-term maintenance of ER expression, function, and response in vitro remains a significant challenge. Here, we report the development of an ER+ breast tumor organoid medium (BTOM-ER) that conserves ER expression, estrogen responsiveness, and dependence, as well as sensitivity to endocrine therapy of ER+ patient-derived xenograft organoids (PDXO). Our findings demonstrate the utility of subtype-specific culture conditions that better mimic the characteristics of the breast epithelial biology and microenvironment, providing a powerful platform for investigating therapy response and disease progression of ER+ breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Receptors, Estrogen/metabolism , Estrogens , Organoids/metabolism , Disease Progression , Tumor Microenvironment
2.
Neuro Oncol ; 25(12): 2287-2301, 2023 12 08.
Article in English | MEDLINE | ID: mdl-37486991

ABSTRACT

BACKGROUND: Medulloblastoma is the most common pediatric brain malignancy. Patients with the Group 3 subtype of medulloblastoma (MB) often exhibit MYC amplification and/or overexpression and have the poorest prognosis. While Group 3 MB is known to be highly dependent on MYC, direct targeting of MYC remains elusive. METHODS: Patient gene expression data were used to identify highly expressed EYA2 in Group 3 MB samples, assess the correlation between EYA2 and MYC, and examine patient survival. Genetic and pharmacological studies were performed on EYA2 in Group 3 derived MB cell models to assess MYC regulation and viability in vitro and in vivo. RESULTS: EYA2 is more highly expressed in Group 3 MB than other MB subgroups and is essential for Group 3 MB growth in vitro and in vivo. EYA2 regulates MYC expression and protein stability in Group 3 MB, resulting in global alterations of MYC transcription. Inhibition of EYA2 tyrosine phosphatase activity, using a novel small molecule inhibitor (NCGC00249987, or 9987), significantly decreases Group 3 MB MYC expression in both flank and intracranial growth in vivo. Human MB RNA-seq data show that EYA2 and MYC are significantly positively correlated, high EYA2 expression is significantly associated with a MYC transcriptional signature, and patients with high EYA2 and MYC expression have worse prognoses than those that do not express both genes at high levels. CONCLUSIONS: Our data demonstrate that EYA2 is a critical regulator of MYC in Group 3 MB and suggest a novel therapeutic avenue to target this highly lethal disease.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Humans , Child , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/metabolism , Cell Line, Tumor , Protein Tyrosine Phosphatases/genetics , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Tyrosine , Nuclear Proteins/genetics , Intracellular Signaling Peptides and Proteins
3.
Nat Commun ; 14(1): 688, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36755019

ABSTRACT

A proper understanding of disease etiology will require longitudinal systems-scale reconstruction of the multitiered architecture of eukaryotic signaling. Here we combine state-of-the-art data acquisition platforms and bioinformatics tools to devise PAMAF, a workflow that simultaneously examines twelve omics modalities, i.e., protein abundance from whole-cells, nucleus, exosomes, secretome and membrane; N-glycosylation, phosphorylation; metabolites; mRNA, miRNA; and, in parallel, single-cell transcriptomes. We apply PAMAF in an established in vitro model of TGFß-induced epithelial to mesenchymal transition (EMT) to quantify >61,000 molecules from 12 omics and 10 timepoints over 12 days. Bioinformatics analysis of this EMT-ExMap resource allowed us to identify; -topological coupling between omics, -four distinct cell states during EMT, -omics-specific kinetic paths, -stage-specific multi-omics characteristics, -distinct regulatory classes of genes, -ligand-receptor mediated intercellular crosstalk by integrating scRNAseq and subcellular proteomics, and -combinatorial drug targets (e.g., Hedgehog signaling and CAMK-II) to inhibit EMT, which we validate using a 3D mammary duct-on-a-chip platform. Overall, this study provides a resource on TGFß signaling and EMT.


Subject(s)
Epithelial-Mesenchymal Transition , Hedgehog Proteins , Epithelial-Mesenchymal Transition/genetics , Hedgehog Proteins/metabolism , Epithelial Cells/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
4.
Cancer Immunol Res ; 10(6): 680-697, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35446942

ABSTRACT

Animal models are critical for the preclinical validation of cancer immunotherapies. Unfortunately, mouse breast cancer models do not faithfully reproduce the molecular subtypes and immune environment of the human disease. In particular, there are no good murine models of estrogen receptor-positive (ER+) breast cancer, the predominant subtype in patients. Here, we show that Nitroso-N-methylurea-induced mammary tumors in outbred Sprague-Dawley rats recapitulate the heterogeneity for mutational profiles, ER expression, and immune evasive mechanisms observed in human breast cancer. We demonstrate the utility of this model for preclinical studies by dissecting mechanisms of response to immunotherapy using combination TGFBR inhibition and PD-L1 blockade. Short-term treatment of early-stage tumors induced durable responses. Gene expression profiling and spatial mapping classified tumors as inflammatory and noninflammatory, and identified IFNγ, T-cell receptor (TCR), and B-cell receptor (BCR) signaling, CD74/MHC II, and epithelium-interacting CD8+ T cells as markers of response, whereas the complement system, M2 macrophage phenotype, and translation in mitochondria were associated with resistance. We found that the expression of CD74 correlated with leukocyte fraction and TCR diversity in human breast cancer. We identified a subset of rat ER+ tumors marked by expression of antigen-processing genes that had an active immune environment and responded to treatment. A gene signature characteristic of these tumors predicted disease-free survival in patients with ER+ Luminal A breast cancer and overall survival in patients with metastatic breast cancer receiving anti-PD-L1 therapy. We demonstrate the usefulness of this preclinical model for immunotherapy and suggest examination to expand immunotherapy to a subset of patients with ER+ disease. See related Spotlight by Roussos Torres, p. 672.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Female , Hormones , Humans , Immunologic Factors , Immunotherapy , Mice , Rats , Rats, Sprague-Dawley , Receptors, Antigen, T-Cell
5.
Cancer Res ; 80(12): 2689-2702, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32341035

ABSTRACT

Metastasis is the major cause of mortality for patients with cancer, and dysregulation of developmental signaling pathways can significantly contribute to the metastatic process. The Sine oculis homeobox homolog 1 (SIX1)/eyes absent (EYA) transcriptional complex plays a critical role in the development of multiple organs and is typically downregulated after development is complete. In breast cancer, aberrant expression of SIX1 has been demonstrated to stimulate metastasis through activation of TGFß signaling and subsequent induction of epithelial-mesenchymal transition (EMT). In addition, SIX1 can induce metastasis via non-cell autonomous means, including activation of GLI-signaling in neighboring tumor cells and activation of VEGFC-induced lymphangiogenesis. Thus, targeting SIX1 would be expected to inhibit metastasis while conferring limited side effects. However, transcription factors are notoriously difficult to target, and thus novel approaches to inhibit their action must be taken. Here we identified a novel small molecule compound, NCGC00378430 (abbreviated as 8430), that reduces the SIX1/EYA2 interaction. 8430 partially reversed transcriptional and metabolic profiles mediated by SIX1 overexpression and reversed SIX1-induced TGFß signaling and EMT. 8430 was well tolerated when delivered to mice and significantly suppressed breast cancer-associated metastasis in vivo without significantly altering primary tumor growth. Thus, we have demonstrated for the first time that pharmacologic inhibition of the SIX1/EYA2 complex and associated phenotypes is sufficient to suppress breast cancer metastasis. SIGNIFICANCE: These findings identify and characterize a novel inhibitor of the SIX1/EYA2 complex that reverses EMT phenotypes suppressing breast cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Homeodomain Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , BRCA1 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , MCF-7 Cells , Mice , Neoplasm Metastasis/prevention & control , Nuclear Proteins/metabolism , Protein Binding/drug effects , Protein Tyrosine Phosphatases/metabolism , RNA-Seq , Signal Transduction/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays
6.
Cancer Res ; 79(4): 720-734, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30606720

ABSTRACT

The capacity for tumor cells to metastasize efficiently is directly linked to their ability to colonize secondary sites. Here we identify Six2, a developmental transcription factor, as a critical regulator of a breast cancer stem cell program that enables metastatic colonization. In several triple-negative breast cancer (TNBC) models, Six2 enhanced the expression of genes associated with embryonic stem cell programs. Six2 directly bound the Sox2 Srr2 enhancer, promoting Sox2 expression and downstream expression of Nanog, which are both key pluripotency factors. Regulation of Sox2 by Six2 enhanced cancer stem cell properties and increased metastatic colonization. Six2 and Sox2 expression correlated highly in breast cancers including TNBC, where a Six2 expression signature was predictive of metastatic burden and poor clinical outcome. Our findings demonstrate that a SIX2/SOX2 axis is required for efficient metastatic colonization, underscoring a key role for stemness factors in outgrowth at secondary sites. SIGNIFICANCE: These findings provide novel mechanistic insight into stemness and the metastatic outgrowth of triple-negative breast cancer cells.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/4/720/F1.large.jpg.


Subject(s)
Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Triple Negative Breast Neoplasms/secondary , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Female , Follow-Up Studies , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplasm Metastasis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Prognosis , SOXB1 Transcription Factors/genetics , Survival Rate , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Nat Commun ; 9(1): 4720, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420638

ABSTRACT

This Article contains an error in Figure 2. In panel a, the second lane of the western blot should have been labelled 'siNT'. A correct version of Figure 2a appears in the Author Correction associated with this Article; the error has not been fixed in the original Article.

8.
Nat Commun ; 8: 15773, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28604738

ABSTRACT

Recent fate-mapping studies concluded that EMT is not required for metastasis of carcinomas. Here we challenge this conclusion by showing that these studies failed to account for possible crosstalk between EMT and non-EMT cells that promotes dissemination of non-EMT cells. In breast cancer models, EMT cells induce increased metastasis of weakly metastatic, non-EMT tumour cells in a paracrine manner, in part by non-cell autonomous activation of the GLI transcription factor. Treatment with GANT61, a GLI1/2 inhibitor, but not with IPI 926, a Smoothened inhibitor, blocks this effect and inhibits growth in PDX models. In human breast tumours, the EMT-transcription factors strongly correlate with activated Hedgehog/GLI signalling but not with the Hh ligands. Our findings indicate that EMT contributes to metastasis via non-cell autonomous effects that activate the Hh pathway. Although all Hh inhibitors may act against tumours with canonical Hh/GLI signalling, only GLI inhibitors would act against non-canonical EMT-induced GLI activation.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Neoplasm Metastasis , Paracrine Communication , Zinc Finger Protein GLI1/metabolism , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Mice, Inbred NOD , Mice, SCID , Pyridines/pharmacology , Pyrimidines/pharmacology , Tumor Microenvironment , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1/antagonists & inhibitors
9.
Nat Commun ; 6: 10077, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26687066

ABSTRACT

TP53 is mutated in 50% of all cancers, and its function is often compromised in cancers where it is not mutated. Here we demonstrate that the pro-tumorigenic/metastatic Six1 homeoprotein decreases p53 levels through a mechanism that does not involve the negative regulator of p53, MDM2. Instead, Six1 regulates p53 via a dual mechanism involving upregulation of microRNA-27a and downregulation of ribosomal protein L26 (RPL26). Mutation analysis confirms that RPL26 inhibits miR-27a binding and prevents microRNA-mediated downregulation of p53. The clinical relevance of this interaction is underscored by the finding that Six1 expression strongly correlates with decreased RPL26 across numerous tumour types. Importantly, we find that Six1 expression leads to marked resistance to therapies targeting the p53-MDM2 interaction. Thus, we identify a competitive mechanism of p53 regulation, which may have consequences for drugs aimed at reinstating p53 function in tumours.


Subject(s)
Down-Regulation , Homeodomain Proteins/metabolism , MicroRNAs/genetics , Neoplasms/genetics , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , MicroRNAs/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL