Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell Death Dis ; 15(4): 296, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670941

ABSTRACT

Genes of the Sprouty family (Spry1-4) restrain signaling by certain receptor tyrosine kinases. Consequently, these genes participate in several developmental processes and function as tumor suppressors in adult life. Despite these important roles, the biology of this family of genes still remains obscure. Here we show that Sprouty proteins are general mediators of cellular senescence. Induction of cellular senescence by several triggers in vitro correlates with upregulation of Sprouty protein levels. More importantly, overexpression of Sprouty genes is sufficient to cause premature cellular senescence, via a conserved N-terminal tyrosine (Tyrosine 53 of Sprouty1). Accordingly, fibroblasts from knockin animals lacking that tyrosine escape replicative senescence. In vivo, heterozygous knockin mice display delayed induction of cellular senescence during cutaneous wound healing and upon chemotherapy-induced cellular senescence. Unlike other functions of this family of genes, induction of cellular senescence appears to be independent of activation of the ERK1/2 pathway. Instead, we show that Sprouty proteins induce cellular senescence upstream of the p38 pathway in these in vitro and in vivo paradigms.


Subject(s)
Cellular Senescence , Fibroblasts , Membrane Proteins , Animals , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Fibroblasts/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Wound Healing
2.
Adv Sci (Weinh) ; : e2204211, 2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36373729

ABSTRACT

Clinical management of endometrial cancer (EC) is handicapped by the limited availability of second line treatments and bona fide molecular biomarkers to predict recurrence. These limitations have hampered the treatment of these patients, whose survival rates have not improved over the last four decades. The advent of coordinated studies such as The Cancer Genome Atlas Uterine Corpus Endometrial Carcinoma (TCGA_UCEC) has partially solved this issue, but the lack of proper experimental systems still represents a bottleneck that precludes translational studies from successful clinical testing in EC patients. Within this context, the first study reporting the generation of a collection of endometrioid-EC-patient-derived orthoxenograft (PDOX) mouse models is presented that is believed to overcome these experimental constraints and pave the way toward state-of-the-art precision medicine in EC. The collection of primary tumors and derived PDOXs is characterized through an integrative approach based on transcriptomics, mutational profiles, and morphological analysis; and it is demonstrated that EC tumors engrafted in the mouse uterus retain the main molecular and morphological features from analogous tumor donors. Finally, the molecular properties of these tumors are harnessed to assess the therapeutic potential of trastuzumab, a human epidermal growth factor receptor 2 (HER2) inhibitor with growing interest in EC, using patient-derived organotypic multicellular tumor spheroids and in vivo experiments.

3.
J Exp Clin Cancer Res ; 41(1): 285, 2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36163066

ABSTRACT

BACKGROUND: Gasdermin B (GSDMB) over-expression promotes poor prognosis and aggressive behavior in HER2 breast cancer by increasing resistance to therapy. Decoding the molecular mechanism of GSDMB-mediated drug resistance is crucial to identify novel effective targeted treatments for HER2/GSDMB aggressive tumors. METHODS: Different in vitro approaches (immunoblot, qRT-PCR, flow cytometry, proteomic analysis, immunoprecipitation, and confocal/electron microscopy) were performed in HER2 breast and gastroesophageal carcinoma cell models. Results were then validated using in vivo preclinical animal models and analyzing human breast and gastric cancer samples. RESULTS: GSDMB up-regulation renders HER2 cancer cells more resistant to anti-HER2 agents by promoting protective autophagy. Accordingly, the combination of lapatinib with the autophagy inhibitor chloroquine increases the therapeutic response of GSDMB-positive cancers in vitro and in zebrafish and mice tumor xenograft in vivo models. Mechanistically, GSDMB N-terminal domain interacts with the key components of the autophagy machinery LC3B and Rab7, facilitating the Rab7 activation during pro-survival autophagy in response to anti-HER2 therapies. Finally, we validated these results in clinical samples where GSDMB/Rab7/LC3B co-expression associates significantly with relapse in HER2 breast and gastric cancers. CONCLUSION: Our findings uncover for the first time a functional link between GSDMB over-expression and protective autophagy in response to HER2-targeted therapies. GSDMB behaves like an autophagy adaptor and plays a pivotal role in modulating autophagosome maturation through Rab7 activation. Finally, our results provide a new and accessible therapeutic approach for HER2/GSDMB + cancers with adverse clinical outcome.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Animals , Autophagy , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Chloroquine/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Lapatinib/pharmacology , Mice , Neoplasm Recurrence, Local , Proteomics , Receptor, ErbB-2/genetics , Zebrafish
4.
JCI Insight ; 7(14)2022 07 22.
Article in English | MEDLINE | ID: mdl-35737463

ABSTRACT

The ribosomal protein S6 kinase 1 (S6K1) is a relevant effector downstream of the mammalian target of rapamycin complex 1 (mTORC1), best known for its role in the control of lipid homeostasis. Consistent with this, mice lacking the S6k1 gene have a defect in their ability to induce the commitment of fat precursor cells to the adipogenic lineage, which contributes to a significant reduction of fat mass. Here, we assess the therapeutic blockage of S6K1 in diet-induced obese mice challenged with LY2584702 tosylate, a specific oral S6K1 inhibitor initially developed for the treatment of solid tumors. We show that diminished S6K1 activity hampers fat mass expansion and ameliorates dyslipidemia and hepatic steatosis, while modifying transcriptome-wide gene expression programs relevant for adipose and liver function. Accordingly, decreased mTORC1 signaling in fat (but increased in the liver) segregated with defective epithelial-mesenchymal transition and the impaired expression of Cd36 (coding for a fatty acid translocase) and Lgals1 (Galectin 1) in both tissues. All these factors combined align with reduced adipocyte size and improved lipidomic signatures in the liver, while hepatic steatosis and hypertriglyceridemia were improved in treatments lasting either 3 months or 6 weeks.


Subject(s)
Fatty Liver , TOR Serine-Threonine Kinases , Animals , Diet , Fatty Liver/drug therapy , Fatty Liver/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
5.
Adv Sci (Weinh) ; 9(4): e2104759, 2022 02.
Article in English | MEDLINE | ID: mdl-34898027

ABSTRACT

The H19X-encoded miR-424(322)/503 cluster regulates multiple cellular functions. Here, it is reported for the first time that it is also a critical linchpin of fat mass expansion. Deletion of this miRNA cluster in mice results in obesity, while increasing the pool of early adipocyte progenitors and hypertrophied adipocytes. Complementary loss and gain of function experiments and RNA sequencing demonstrate that miR-424(322)/503 regulates a conserved genetic program involved in the differentiation and commitment of white adipocytes. Mechanistically, it is demonstrated that miR-424(322)/503 targets γ-Synuclein (SNCG), a factor that mediates this program rearrangement by controlling metabolic functions in fat cells, allowing adipocyte differentiation and adipose tissue enlargement. Accordingly, diminished miR-424(322) in mice and obese humans co-segregate with increased SNCG in fat and peripheral blood as mutually exclusive features of obesity, being normalized upon weight loss. The data unveil a previously unknown regulatory mechanism of fat mass expansion tightly controlled by the miR-424(322)/503 through SNCG.


Subject(s)
Adipose Tissue/metabolism , Cell Differentiation , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , gamma-Synuclein/metabolism , Adipogenesis , Animals , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Proteins/genetics , gamma-Synuclein/genetics
6.
Cancers (Basel) ; 13(24)2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34944822

ABSTRACT

About 70% of advanced-stage prostate cancer (PCa) patients will experience bone metastasis, which severely affects patients' quality of life and progresses to lethal PCa in most cases. Hence, understanding the molecular heterogeneity of PCa cell populations and the signaling pathways associated with bone tropism is crucial. For this purpose, we generated an animal model with high penetrance to metastasize to bone using an intracardiac percutaneous injection of PC3 cells to identify PCa metastasis-promoting factors. Using genomic high-throughput analysis we identified a miRNA signature involved in bone metastasis that also presents potential as a biomarker of PCa progression in human samples. In particular, the downregulation of miR-135b favored the incidence of bone metastases by significantly increasing PCa cells' migratory capacity. Moreover, the PLAG1, JAKMIP2, PDGFA, and VTI1b target genes were identified as potential mediators of miR-135b's role in the dissemination to bone. In this study, we provide a genomic signature involved in PCa bone growth, contributing to a better understanding of the mechanisms responsible for this process. In the future, our results could ultimately translate into promising new therapeutic targets for the treatment of lethal PCa.

7.
EMBO Rep ; 22(12): e53201, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34633138

ABSTRACT

During the female lifetime, the expansion of the epithelium dictated by the ovarian cycles is supported by a transient increase in the mammary epithelial stem cell population (MaSCs). Notably, activation of Wnt/ß-catenin signaling is an important trigger for MaSC expansion. Here, we report that the miR-424/503 cluster is a modulator of canonical Wnt signaling in the mammary epithelium. We show that mammary tumors of miR-424(322)/503-depleted mice exhibit activated Wnt/ß-catenin signaling. Importantly, we show a strong association between miR-424/503 deletion and breast cancers with high levels of Wnt/ß-catenin signaling. Moreover, miR-424/503 cluster is required for Wnt-mediated MaSC expansion induced by the ovarian cycles. Lastly, we show that miR-424/503 exerts its function by targeting two binding sites at the 3'UTR of the LRP6 co-receptor and reducing its expression. These results unveil an unknown link between the miR-424/503, regulation of Wnt signaling, MaSC fate, and tumorigenesis.


Subject(s)
Epithelium , Low Density Lipoprotein Receptor-Related Protein-6 , Mammary Glands, Animal/cytology , MicroRNAs , Wnt Signaling Pathway , Animals , Breast Neoplasms , Carcinogenesis , Cell Line, Tumor , Epithelial Cells/cytology , Epithelium/metabolism , Female , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Menstrual Cycle , Mice , MicroRNAs/genetics , Stem Cells/cytology
8.
Mol Oncol ; 12(7): 1061-1076, 2018 06.
Article in English | MEDLINE | ID: mdl-29683256

ABSTRACT

The neuregulins (NRGs) represent a large family of membrane-anchored growth factors, whose deregulation may contribute to the pathogenesis of several tumors. In fact, targeting of NRG-activated pathways has demonstrated clinical benefit. To improve the efficacy of anti-NRG therapies, it is essential to gain insights into the regions of NRGs that favor their pro-oncogenic properties. Here, we have addressed the protumorigenic impact of different NRG domains. To do this, deletion mutants affecting different NRG domains were expressed in 293 and MCF7 cells. Of the five forms studied, only the wild-type and a mutant lacking the Ig-like domain (NRGΔIg ) were properly sorted to the plasma membrane. Both forms were released as soluble forms to the culture media. However, the mutant NRGΔIg failed to efficiently activate HER2 and HER3 receptors, signaling pathways, and cell proliferation when compared to wild-type NRG. Treatment with trastuzumab, a humanized antibody used in the breast cancer clinic, inhibited the constitutive activation of HER2, HER3, and downstream signaling in MCF7 cells constitutively expressing wild-type NRG. In contrast, this treatment had a marginal effect on MCF7-NRGΔIg cells. This study demonstrates that the Ig-like region of NRGs exerts an important role in their capability to activate ErbB/HER receptors and mitogenic responses. Strategies aimed at targeting NRGs should consider that fact to improve neutralization of the pro-oncogenic properties of NRGs.


Subject(s)
Immunoglobulin Domains , Neuregulins/chemistry , Neuregulins/metabolism , Receptor, ErbB-2/metabolism , Cell Membrane/metabolism , Cell Proliferation , Humans , MCF-7 Cells , Mutant Proteins/metabolism , Signal Transduction , Solubility , Structure-Activity Relationship , Trastuzumab
9.
Cancer Discov ; 8(5): 582-599, 2018 05.
Article in English | MEDLINE | ID: mdl-29510988

ABSTRACT

High-risk neuroblastomas show a paucity of recurrent somatic mutations at diagnosis. As a result, the molecular basis for this aggressive phenotype remains elusive. Recent progress in regulatory network analysis helped us elucidate disease-driving mechanisms downstream of genomic alterations, including recurrent chromosomal alterations. Our analysis identified three molecular subtypes of high-risk neuroblastomas, consistent with chromosomal alterations, and identified subtype-specific master regulator proteins that were conserved across independent cohorts. A 10-protein transcriptional module-centered around a TEAD4-MYCN positive feedback loop-emerged as the regulatory driver of the high-risk subtype associated with MYCN amplification. Silencing of either gene collapsed MYCN-amplified (MYCNAmp) neuroblastoma transcriptional hallmarks and abrogated viability in vitro and in vivo Consistently, TEAD4 emerged as a robust prognostic marker of poor survival, with activity independent of the canonical Hippo pathway transcriptional coactivators YAP and TAZ. These results suggest novel therapeutic strategies for the large subset of MYCN-deregulated neuroblastomas.Significance: Despite progress in understanding of neuroblastoma genetics, little progress has been made toward personalized treatment. Here, we present a framework to determine the downstream effectors of the genetic alterations sustaining neuroblastoma subtypes, which can be easily extended to other tumor types. We show the critical effect of disrupting a 10-protein module centered around a YAP/TAZ-independent TEAD4-MYCN positive feedback loop in MYCNAmp neuroblastomas, nominating TEAD4 as a novel candidate for therapeutic intervention. Cancer Discov; 8(5); 582-99. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 517.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Muscle Proteins/genetics , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Acyltransferases , Cell Cycle Proteins , Cell Line, Tumor , Computational Biology/methods , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Humans , Muscle Proteins/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neoplasm Staging , Neuroblastoma/diagnosis , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA Interference , TEA Domain Transcription Factors , Transcription Factors/metabolism , Transcriptional Activation
10.
Genes Dev ; 31(6): 553-566, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28404630

ABSTRACT

The female mammary gland is a very dynamic organ that undergoes continuous tissue remodeling during adulthood. Although it is well established that the number of menstrual cycles and pregnancy (in this case transiently) increase the risk of breast cancer, the reasons are unclear. Growing clinical and experimental evidence indicates that improper involution plays a role in the development of this malignancy. Recently, we described the miR-424(322)/503 cluster as an important regulator of mammary epithelial involution after pregnancy. Here, through the analysis of ∼3000 primary tumors, we show that miR-424(322)/503 is commonly lost in a subset of aggressive breast cancers and describe the genetic aberrations that inactivate its expression. Furthermore, through the use of a knockout mouse model, we demonstrate for the first time that loss of miR-424(322)/503 promotes breast tumorigenesis in vivo. Remarkably, we found that loss of miR-424(322)/503 promotes chemoresistance due to the up-regulation of two of its targets: BCL-2 and insulin-like growth factor-1 receptor (IGF1R). Importantly, targeted therapies blocking the aberrant activity of these targets restore sensitivity to chemotherapy. Overall, our studies reveal miR-424(322)/503 as a tumor suppressor in breast cancer and provide a link between mammary epithelial involution, tumorigenesis, and the phenomenon of chemoresistance.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Deletion , Genes, Tumor Suppressor , Humans , Mammary Neoplasms, Experimental/genetics , Mice , Pregnancy , Pregnancy Complications, Neoplastic/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , cdc25 Phosphatases/genetics
12.
Autophagy ; 13(3): 608-624, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28055301

ABSTRACT

Targeted therapies in endometrial cancer (EC) using kinase inhibitors rarely result in complete tumor remission and are frequently challenged by the appearance of refractory cell clones, eventually resulting in disease relapse. Dissecting adaptive mechanisms is of vital importance to circumvent clinical drug resistance and improve the efficacy of targeted agents in EC. Sorafenib is an FDA-approved multitarget tyrosine and serine/threonine kinase inhibitor currently used to treat hepatocellular carcinoma, advanced renal carcinoma and radioactive iodine-resistant thyroid carcinoma. Unfortunately, sorafenib showed very modest effects in a multi-institutional phase II trial in advanced uterine carcinoma patients. Here, by leveraging RNA-sequencing data from the Cancer Cell Line Encyclopedia and cell survival studies from compound-based high-throughput screenings we have identified the lysosomal pathway as a potential compartment involved in the resistance to sorafenib. By performing additional functional biology studies we have demonstrated that this resistance could be related to macroautophagy/autophagy. Specifically, our results indicate that sorafenib triggers a mechanistic MAPK/JNK-dependent early protective autophagic response in EC cells, providing an adaptive response to therapeutic stress. By generating in vivo subcutaneous EC cell line tumors, lung metastatic assays and primary EC orthoxenografts experiments, we demonstrate that targeting autophagy enhances sorafenib cytotoxicity and suppresses tumor growth and pulmonary metastasis progression. In conclusion, sorafenib induces the activation of a protective autophagic response in EC cells. These results provide insights into the unopposed resistance of advanced EC to sorafenib and highlight a new strategy for therapeutic intervention in recurrent EC.


Subject(s)
Autophagy , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Molecular Targeted Therapy , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Disease Progression , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/ultrastructure , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Female , Humans , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Sorafenib , Xenograft Model Antitumor Assays
13.
Oncotarget ; 7(20): 28976-88, 2016 May 17.
Article in English | MEDLINE | ID: mdl-26716514

ABSTRACT

Survival for lung cancer patients remains dismal and is largely attributed to treatment resistance. To identify novel target genes the modulation of which could modify platinum resistance, we performed a high-throughput RNAi screen and identified Yes-associated protein (YAP1), a transcription coactivator and a known oncogene, as a potential actionable candidate. YAP1 ablation significantly improved sensitivities not only to cisplatin but also to ionizing radiation, both of which are DNA-damaging interventions, in non-small cell lung cancer (NSCLC) cells. Overall YAP1 was expressed in 75% of NSCLC specimens, whereas nuclear YAP1 which is the active form was present in 45% of 124 resected NSCLC. Interestingly, EGFR-mutated or KRAS-mutated NSCLC were associated with higher nuclear YAP1 staining in comparison to EGFR/KRAS wild-type. Relevantly, YAP1 downregulation improved sensitivity to erlotinib, an EGFR inhibitor. A pharmacological inhibitor of YAP1 signaling, verteporfin also synergized with cisplatin, radiation and erlotinib in NSCLC cells by potentiating cisplatin and radiation-related double-stranded breaks and decreasing expression of YAP1 and EGFR. Taken together, our study is the first to indicate the potential role of YAP1 as a common modulator of resistance mechanisms and a potential novel, actionable target that can improve responses to platinum, radiation and EGFR-targeted therapy in lung cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/pathology , Phosphoproteins/genetics , Radiation Tolerance/genetics , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/genetics , Transcription Factors , YAP-Signaling Proteins
14.
Breast Cancer Res ; 17(1): 149, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26643555

ABSTRACT

INTRODUCTION: Inflammatory breast cancer (IBC) is the most lethal form of breast cancers with a 5-year survival rate of only 40 %. Despite its lethality, IBC remains poorly understood which has greatly limited its therapeutic management. We thus decided to utilize an integrative functional genomic strategy to identify the Achilles' heel of IBC cells. METHODS: We have pioneered the development of genetic tools as well as experimental and analytical strategies to perform RNAi-based loss-of-function studies at a genome-wide level. Importantly, we and others have demonstrated that these functional screens are able to identify essential functions linked to certain cancer phenotypes. Thus, we decided to use this approach to identify IBC specific sensitivities. RESULTS: We identified and validated HDAC6 as a functionally necessary gene to maintain IBC cell viability, while being non-essential for other breast cancer subtypes. Importantly, small molecule inhibitors for HDAC6 already exist and are in clinical trials for other tumor types. We thus demonstrated that Ricolinostat (ACY1215), a leading HDAC6 inhibitor, efficiently controls IBC cell proliferation both in vitro and in vivo. Critically, functional HDAC6 dependency is not associated with genomic alterations at its locus and thus represents a non-oncogene addiction. Despite HDAC6 not being overexpressed, we found that its activity is significantly higher in IBC compared to non-IBC cells, suggesting a possible rationale supporting the observed dependency. CONCLUSION: Our finding that IBC cells are sensitive to HDAC6 inhibition provides a foundation to rapidly develop novel, efficient, and well-tolerated targeted therapy strategies for IBC patients.


Subject(s)
Histone Deacetylases/metabolism , Inflammatory Breast Neoplasms/enzymology , Cell Line, Tumor , Cell Survival , Female , Gene Expression , Gene Knockdown Techniques , Gene Ontology , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Inflammatory Breast Neoplasms/pathology
15.
Genes Dev ; 29(15): 1631-48, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26227964

ABSTRACT

HER2-positive (HER2(+)) breast adenocarcinomas are a heterogeneous group in which hormone receptor (HR) status influences therapeutic decisions and patient outcome. By combining genome-wide RNAi screens with regulatory network analysis, we identified STAT3 as a critically activated master regulator of HR(-)/HER2(+) tumors, eliciting tumor dependency in these cells. Mechanistically, HR(-)/HER2(+) cells secrete high levels of the interleukin-6 (IL-6) cytokine, inducing the activation of STAT3, which in turn promotes a second autocrine stimulus to increase S100A8/9 complex (calprotectin) production and secretion. Increased calprotectin levels activate signaling pathways involved in proliferation and resistance. Importantly, we demonstrated that inhibition of the IL-6-Janus kinase 2 (JAK2)-STAT3-calprotectin axis with FDA-approved drugs, alone and in combination with HER2 inhibitors, reduced the tumorigenicity of HR(-)/HER2(+) breast cancers, opening novel targeted therapeutic opportunities.


Subject(s)
Breast Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic , STAT3 Transcription Factor/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Carcinogenesis/genetics , Cell Line, Tumor , Cell Survival/genetics , Female , Genome-Wide Association Study , Heterografts , Humans , Interleukin-6/metabolism , Janus Kinase 2/metabolism , Mice , Mice, SCID , Quinolines/pharmacology , Quinolones , RNA Interference , STAT3 Transcription Factor/genetics
16.
Oncotarget ; 6(5): 3292-305, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25669969

ABSTRACT

MYC deregulation is a driver of many human cancers. Altering the balance of MYC protein levels at the level of transcription, protein stability, or turnover is sufficient to transform cells to a tumorigenic phenotype. While direct targeting of MYC is difficult, specific genetic vulnerabilities of MYC-deregulated cells could be exploited to selectively inhibit their growth. Using a genome-wide shRNA screen, we identified 78 candidate genes, which are required for survival of human mammary epithelial cells with elevated MYC levels. Among the candidates, we validated and characterized FBXW7, a component of the SCF-like ubiquitin ligase complex that targets MYC for proteasomal degradation. Down-regulation of FBXW7 leads to synergistic accumulation of cellular and active chromatin-bound MYC, while protein levels of other FBXW7 targets appear unaffected. Over a four-week time course, continuous FBXW7 down-regulation and MYC activation together cause an accumulation of cells in S-phase and G2/M-phase of the cell cycle. Under these conditions, we also observe elevated chromatin-bound levels of CDC45, suggesting increased DNA replication stress. Consistent with these results, FBXW7 down-regulation alone decreases the survival of T47D breast cancer cells. These results establish that FBXW7 down-regulation is synthetic lethal with MYC, and that MYC is a critical target of FBXW7 in breast epithelial cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Epithelial Cells/metabolism , F-Box Proteins/metabolism , Mammary Glands, Human/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin-Protein Ligases/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chromatin Assembly and Disassembly , Epithelial Cells/pathology , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mammary Glands, Human/pathology , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , S Phase Cell Cycle Checkpoints , Signal Transduction , Time Factors , Transfection , Ubiquitin-Protein Ligases/genetics
17.
Cancer Cell ; 27(2): 223-39, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25670080

ABSTRACT

Elucidating the determinants of aggressiveness in lethal prostate cancer may stimulate therapeutic strategies that improve clinical outcomes. We used experimental models and clinical databases to identify GATA2 as a regulator of chemotherapy resistance and tumorigenicity in this context. Mechanistically, direct upregulation of the growth hormone IGF2 emerged as a mediator of the aggressive properties regulated by GATA2. IGF2 in turn activated IGF1R and INSR as well as a downstream polykinase program. The characterization of this axis prompted a combination strategy whereby dual IGF1R/INSR inhibition restored the efficacy of chemotherapy and improved survival in preclinical models. These studies reveal a GATA2-IGF2 aggressiveness axis in lethal prostate cancer and identify a therapeutic opportunity in this challenging disease.


Subject(s)
GATA2 Transcription Factor/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Animals , Antigens, CD/genetics , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Humans , Insulin-Like Growth Factor II/genetics , Male , Mice , Prostatic Neoplasms, Castration-Resistant/pathology , Receptor, Insulin/genetics , Signal Transduction , Xenograft Model Antitumor Assays
18.
Cell ; 159(6): 1461-75, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25433701

ABSTRACT

Identifying driver genes in cancer remains a crucial bottleneck in therapeutic development and basic understanding of the disease. We developed Helios, an algorithm that integrates genomic data from primary tumors with data from functional RNAi screens to pinpoint driver genes within large recurrently amplified regions of DNA. Applying Helios to breast cancer data identified a set of candidate drivers highly enriched with known drivers (p < 10(-14)). Nine of ten top-scoring Helios genes are known drivers of breast cancer, and in vitro validation of 12 candidates predicted by Helios found ten conferred enhanced anchorage-independent growth, demonstrating Helios's exquisite sensitivity and specificity. We extensively characterized RSF-1, a driver identified by Helios whose amplification correlates with poor prognosis, and found increased tumorigenesis and metastasis in mouse models. We have demonstrated a powerful approach for identifying driver genes and how it can yield important insights into cancer.


Subject(s)
Algorithms , Breast Neoplasms/genetics , Animals , Bayes Theorem , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Copy Number Variations , Female , Genome-Wide Association Study , Humans , Mice, Inbred NOD , Mice, SCID , RNA Interference
19.
Mol Cell Biol ; 34(23): 4216-31, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25266660

ABSTRACT

Recently, we demonstrated that the microRNA 424(322)/503 [miR-424(322)/503] cluster is transcriptionally controlled by transforming growth factor ß (TGF-ß) in the mammary epithelium. Induction of this microRNA cluster impacts mammary epithelium fate by regulating apoptosis and insulin-like growth factor 1 (IGF1) signaling. Here, we expanded our finding to demonstrate that miR-424(322)/503 is an integral component of the cell cycle arrest mediated by TGF-ß. Mechanistically, we showed that after TGF-ß exposure, increased levels of miR-424(322)/503 reduce the expression of the cell cycle regulator CDC25A. miR-424(322)/503-dependent posttranscriptional downregulation of CDC25A cooperates with previously described transcriptional repression of the CDC25A promoter and proteasome-mediated degradation to reduce the levels of CDC25A expression and to induce cell cycle arrest. We also provide evidence that the TGF-ß/miR-424(322)/503 axis is part of the mechanism that regulates the proliferation of hormone receptor-positive (HR(+)) mammary epithelial cells in vivo.


Subject(s)
Mammary Glands, Human/growth & development , MicroRNAs/genetics , Transforming Growth Factor beta/metabolism , cdc25 Phosphatases/biosynthesis , Animals , Apoptosis/genetics , Cell Line , Cell Proliferation/genetics , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , G1 Phase Cell Cycle Checkpoints/genetics , Humans , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Glands, Human/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , Promoter Regions, Genetic , Pyrazoles/pharmacology , Pyrroles/pharmacology , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Transcription, Genetic , Transforming Growth Factor beta/antagonists & inhibitors , cdc25 Phosphatases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...