Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Nat Commun ; 14(1): 4313, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37463901

ABSTRACT

Metastatic breast-cancer is a major cause of death in women worldwide, yet the relationship between oncogenic drivers that promote metastatic versus primary cancer is still contentious. To elucidate this relationship in treatment-naive animals, we hereby describe mammary-specific transposon-mutagenesis screens in female mice together with loss-of-function Rb, which is frequently inactivated in breast-cancer. We report gene-centric common insertion-sites (gCIS) that are enriched in primary-tumors, in metastases or shared by both compartments. Shared-gCIS comprise a major MET-RAS network, whereas metastasis-gCIS form three additional hubs: Rho-signaling, Ubiquitination and RNA-processing. Pathway analysis of four clinical cohorts with paired primary-tumors and metastases reveals similar organization in human breast-cancer with subtype-specific shared-drivers (e.g. RB1-loss, TP53-loss, high MET, RAS, ER), primary-enriched (EGFR, TGFß and STAT3) and metastasis-enriched (RHO, PI3K) oncogenic signaling. Inhibitors of RB1-deficiency or MET plus RHO-signaling cooperate to block cell migration and drive tumor cell-death. Thus, targeting shared- and metastasis- but not primary-enriched derivers offers a rational avenue to prevent metastatic breast-cancer.


Subject(s)
Breast Neoplasms , Female , Humans , Animals , Mice , Breast Neoplasms/pathology , Signal Transduction , Neoplasm Metastasis
2.
Development ; 149(19)2022 10 01.
Article in English | MEDLINE | ID: mdl-36205077

ABSTRACT

Notch3 promotes mammary luminal cell specification and forced Notch3 activation can induce mammary tumor formation. However, recent studies suggest a tumor-suppressive role for Notch3. Here, we report on Notch3 expression and functional analysis in the mouse mammary gland. Notch3 is expressed in the luminal compartment throughout mammary gland development, but switches to basal cells with initiation of post-lactational involution. Deletion of Notch3 caused a decrease of Notch activation in luminal cells and diminished luminal progenitors at puberty, as well as reduced alveolar progenitors during pregnancy. Parous Notch3-/- mammary glands developed hyperplasia with accumulation of CD24hiCD49flo cells, some of which progressed to invasive tumors with luminal features. Notch3 deletion abolished Notch activation in basal cells during involution, accompanied by altered apoptosis and reduced brown adipocytes, leading to expansion of parity-identified mammary epithelial cells (PI-MECs). Interestingly, the postpartum microenvironment is required for the stem cell activity of Notch3-/- PI-MECs. Finally, high expression of NOTCH3 is associated with prolonged survival in patients with luminal breast cancer. These results highlight an unexpected tumor-suppressive function for Notch3 in the parous mammary gland through restriction of PI-MEC expansion.


Subject(s)
Epithelial Cells , Mammary Glands, Animal , Animals , Epithelial Cells/metabolism , Female , Lactation , Mice , Mice, Transgenic , Pregnancy , Stem Cells
3.
Cancer Discov ; 12(12): 2930-2953, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36108220

ABSTRACT

Systematically investigating the scores of genes mutated in cancer and discerning disease drivers from inconsequential bystanders is a prerequisite for precision medicine but remains challenging. Here, we developed a somatic CRISPR/Cas9 mutagenesis screen to study 215 recurrent "long-tail" breast cancer genes, which revealed epigenetic regulation as a major tumor-suppressive mechanism. We report that components of the BAP1 and COMPASS-like complexes, including KMT2C/D, KDM6A, BAP1, and ASXL1/2 ("EpiDrivers"), cooperate with PIK3CAH1047R to transform mouse and human breast epithelial cells. Mechanistically, we find that activation of PIK3CAH1047R and concomitant EpiDriver loss triggered an alveolar-like lineage conversion of basal mammary epithelial cells and accelerated formation of luminal-like tumors, suggesting a basal origin for luminal tumors. EpiDriver mutations are found in ∼39% of human breast cancers, and ∼50% of ductal carcinoma in situ express casein, suggesting that lineage infidelity and alveogenic mimicry may significantly contribute to early steps of breast cancer etiology. SIGNIFICANCE: Infrequently mutated genes comprise most of the mutational burden in breast tumors but are poorly understood. In vivo CRISPR screening identified functional tumor suppressors that converged on epigenetic regulation. Loss of epigenetic regulators accelerated tumorigenesis and revealed lineage infidelity and aberrant expression of alveogenesis genes as potential early events in tumorigenesis. This article is highlighted in the In This Issue feature, p. 2711.


Subject(s)
Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Humans , Mice , Animals , Female , Breast Neoplasms/pathology , Epigenesis, Genetic , Neoplasm Recurrence, Local/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Cell Transformation, Neoplastic/genetics
4.
Nat Commun ; 12(1): 5238, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34475389

ABSTRACT

The most common events in breast cancer (BC) involve chromosome arm losses and gains. Here we describe identification of 1089 gene-centric common insertion sites (gCIS) from transposon-based screens in 8 mouse models of BC. Some gCIS are driver-specific, others driver non-specific, and still others associated with tumor histology. Processes affected by driver-specific and histology-specific mutations include well-known cancer pathways. Driver non-specific gCIS target the Mediator complex, Ca++ signaling, Cyclin D turnover, RNA-metabolism among other processes. Most gCIS show single allele disruption and many map to genomic regions showing high-frequency hemizygous loss in human BC. Two gCIS, Nf1 and Trps1, show synthetic haploinsufficient tumor suppressor activity. Many gCIS act on the same pathway responsible for tumor initiation, thereby selecting and sculpting just enough and just right signaling. These data highlight ~1000 genes with predicted conditional haploinsufficient tumor suppressor function and the potential to promote chromosome arm loss in BC.


Subject(s)
Breast Neoplasms/genetics , Loss of Heterozygosity/genetics , Animals , Breast Neoplasms/pathology , Cell Transformation, Neoplastic , DNA Transposable Elements/genetics , Female , Genes, Tumor Suppressor , Humans , Mice , Mutagenesis, Insertional , Neoplasms, Experimental , Signal Transduction
5.
Front Cell Dev Biol ; 9: 645386, 2021.
Article in English | MEDLINE | ID: mdl-33996804

ABSTRACT

Epigenetic and chromatin regulation of craniofacial development remains poorly understood. Ankyrin Repeat Domain 11 (ANKRD11) is a chromatin regulator that has previously been shown to control neural stem cell fates via modulation of histone acetylation. ANKRD11 gene variants, or microdeletions of the 16q24.3 chromosomal region encompassing the ANKRD11 gene, cause KBG syndrome, a rare autosomal dominant congenital disorder with variable neurodevelopmental and craniofacial involvement. Craniofacial abnormalities include a distinct facial gestalt, delayed bone age, tooth abnormalities, delayed fontanelle closure, and frequently cleft or submucosal palate. Despite this, the dramatic phenotype and precise role of ANKRD11 in embryonic craniofacial development remain unexplored. Quantitative analysis of 3D images of KBG syndromic subjects shows an overall reduction in the size of the middle and lower face. Here, we report that mice with heterozygous deletion of Ankrd11 in neural crest cells (Ankrd11nchet) display a mild midfacial hypoplasia including reduced midfacial width and a persistent open fontanelle, both of which mirror KBG syndrome patient facial phenotypes. Mice with a homozygous Ankrd11 deletion in neural crest cells (Ankrd11ncko) die at birth. They show increased severity of several clinical manifestations described for KBG syndrome, such as cleft palate, retrognathia, midfacial hypoplasia, and reduced calvarial growth. At E14.5, Ankrd11 expression in the craniofacial complex is closely associated with developing bony structures, while expression at birth is markedly decreased. Conditional deletion of Ankrd11 leads to a reduction in ossification of midfacial bones, with several ossification centers failing to expand and/or fuse. Intramembranous bones show features of delayed maturation, with bone remodeling severely curtailed at birth. Palatal shelves remain hypoplastic at all developmental stages, with a local reduction in proliferation at E13.5. Our study identifies Ankrd11 as a critical regulator of intramembranous ossification and palate development and suggests that Ankrd11nchet and Ankrd11ncko mice may serve as pre-clinical models for KBG syndrome in humans.

6.
EMBO Mol Med ; 13(3): e13807, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33616312

ABSTRACT

Invasive lobular carcinoma (ILC) of the breast is a very common disease. Despite its prevalence, these tumors are relatively understudied. One reason for this is a relative lack of models for ILC. This challenge was addressed by Brisken and colleagues through development of an intraductal injection-based xenograft system for the study of ERα+ breast cancers, including both ILC and more common invasive ductal carcinoma (IDC; Sflomos et al, 2016). In this issue of EMBO Molecular Medicine, the same group have applied intraductal injection-based xenografts to identify novel tumor cell-specific transcriptional signatures in ILC (Sflomos et al, 2021). In doing so they found overexpression of lysyl oxidase-like 1 (LOXL1) to be both responsible for the frequently seen stiff collagen-rich extracellular matrix of lobular breast cancer and essential for their robust growth and metastatic dissemination in vivo, thereby identifying a novel therapeutic target.


Subject(s)
Breast Neoplasms , Carcinoma, Ductal, Breast , Carcinoma, Lobular , Breast , Female , Humans , Retrospective Studies
7.
Cancers (Basel) ; 13(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33578666

ABSTRACT

Metastatic dissemination of cancer cells, their colonization at distal sites, and ultimate disruption of tissue physiology are the root causes of most deaths from solid cancers, particularly in tumor types where the primary lesion can be easily dissected and discarded [...].

8.
Cell Rep ; 25(3): 702-714.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332649

ABSTRACT

CDH1 and PIK3CA are the two most frequently mutated genes in invasive lobular carcinoma (ILC) of the breast. Transcription profiling has identified molecular subtypes for ILC, one of which, immune-related (IR), is associated with gene expression linked to lymphocyte and macrophage infiltration. Here, we report that deletion of Cdh1, together with activation of Pik3ca in mammary epithelium of genetically modified mice, leads to formation of IR-ILC-like tumors with immune cell infiltration, as well as gene expression linked to T-regulatory (Treg) cell signaling and activation of targetable immune checkpoint pathways. Interestingly, these tumors show enhanced Rac1- and Yap-dependent transcription and signaling, as well as sensitivity to PI3K, Rac1, and Yap inhibitors in culture. Finally, high-dimensional immunophenotyping in control mouse mammary gland and IR-ILC tumors by mass cytometry shows dramatic alterations in myeloid and lymphoid populations associated with immune suppression and exhaustion, highlighting the potential for therapeutic intervention via immune checkpoint regulators.


Subject(s)
Cadherins/physiology , Carcinoma, Lobular/pathology , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/pathology , Mutation , Phosphatidylinositol 3-Kinases/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinoma, Lobular/immunology , Carcinoma, Lobular/metabolism , Cell Cycle Proteins/metabolism , Class I Phosphatidylinositol 3-Kinases , Female , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Invasiveness , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transcriptome , YAP-Signaling Proteins , rac GTP-Binding Proteins/metabolism
9.
Cell Rep ; 23(1): 112-126, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617654

ABSTRACT

CDK4/6 inhibitors are effective against cancer cells expressing the tumor suppressor RB1, but not RB1-deficient cells, posing the challenge of how to target RB1 loss. In triple-negative breast cancer (TNBC), RB1 and PTEN are frequently inactivated together with TP53. We performed kinome/phosphatase inhibitor screens on primary mouse Rb/p53-, Pten/p53-, and human RB1/PTEN/TP53-deficient TNBC cell lines and identified CDC25 phosphatase as a common target. Pharmacological or genetic inhibition of CDC25 suppressed growth of RB1-deficient TNBC cells that are resistant to combined CDK4/6 plus CDK2 inhibition. Minimal cooperation was observed in vitro between CDC25 antagonists and CDK1, CDK2, or CDK4/6 inhibitors, but strong synergy with WEE1 inhibition was apparent. In accordance with increased PI3K signaling following long-term CDC25 inhibition, CDC25 and PI3K inhibitors effectively synergized to suppress TNBC growth both in vitro and in xenotransplantation models. These results provide a rationale for the development of CDC25-based therapies for diverse RB1/PTEN/TP53-deficient and -proficient TNBCs.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Triple Negative Breast Neoplasms/drug therapy , cdc25 Phosphatases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/therapeutic use , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
10.
Neuroimage ; 163: 220-230, 2017 12.
Article in English | MEDLINE | ID: mdl-28882630

ABSTRACT

MRI is a powerful modality to detect neuroanatomical differences that result from mutations and treatments. Knowing which genes drive these differences is important in understanding etiology, but candidate genes are often difficult to identify. We tested whether spatial gene expression data from the Allen Brain Institute can be used to inform us about genes that cause neuroanatomical differences. For many single-gene-mutation mouse models, we found that affected neuroanatomy was not strongly associated with the spatial expression of the altered gene and there are specific caveats for each model. However, among models with significant neuroanatomical differences from their wildtype controls, the mutated genes had preferential spatial expression in affected neuroanatomy. In mice exposed to environmental enrichment, candidate genes could be identified by a genome-wide search for genes with preferential spatial expression in the altered neuroanatomical regions. These candidates have functions related to learning and plasticity. We demonstrate that spatial gene expression of single-genes is a poor predictor of altered neuroanatomy, but altered neuroanatomy can identify candidate genes responsible for neuroanatomical phenotypes.


Subject(s)
Brain/anatomy & histology , Animals , Disease Models, Animal , Genetic Association Studies , Mice , Mice, Inbred C57BL , Mutation , Phenotype
11.
Stem Cell Reports ; 7(4): 787-801, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27618721

ABSTRACT

Blood vessels are formed through vasculogenesis, followed by remodeling of the endothelial network through angiogenesis. Many events that occur during embryonic vascular development are recapitulated during adult neoangiogenesis, which is critical to tumor growth and metastasis. Current antiangiogenic tumor therapies, based largely on targeting the vascular endothelial growth factor pathway, show limited clinical benefits, thus necessitating the discovery of alternative targets. Here we report the development of a robust embryonic stem cell-based vascular differentiation assay amenable to small-molecule screens to identify novel modulators of angiogenesis. In this context, RSK and TTK were identified as angiogenic modulators. Inhibition of these pathways inhibited angiogenesis in embryoid bodies and human umbilical vein endothelial cells. Furthermore, inhibition of RSK and TTK reduced tumor growth, vascular density, and improved survival in an in vivo Lewis lung carcinoma mouse model. Our study suggests that RSK and TTK are potential targets for antiangiogenic therapy, and provides an assay system for further pathway screens.


Subject(s)
Blood Vessels/embryology , Blood Vessels/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Ribosomal Protein S6 Kinases/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Discovery , Female , Humans , Mice , Morphogenesis , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Organogenesis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/antagonists & inhibitors
12.
J Clin Invest ; 126(10): 3739-3757, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27571409

ABSTRACT

Triple-negative breast cancer (TNBC) includes basal-like and claudin-low subtypes for which no specific treatment is currently available. Although the retinoblastoma tumor-suppressor gene (RB1) is frequently lost together with TP53 in TNBC, it is not directly targetable. There is thus great interest in identifying vulnerabilities downstream of RB1 that can be therapeutically exploited. Here, we determined that combined inactivation of murine Rb and p53 in diverse mammary epithelial cells induced claudin-low-like TNBC with Met, Birc2/3-Mmp13-Yap1, and Pvt1-Myc amplifications. Gene set enrichment analysis revealed that Rb/p53-deficient tumors showed elevated expression of the mitochondrial protein translation (MPT) gene pathway relative to tumors harboring p53 deletion alone. Accordingly, bioinformatic, functional, and biochemical analyses showed that RB1-E2F complexes bind to MPT gene promoters to regulate transcription and control MPT. Additionally, a screen of US Food and Drug Administration-approved (FDA-approved) drugs identified the MPT antagonist tigecycline (TIG) as a potent inhibitor of Rb/p53-deficient tumor cell proliferation. TIG preferentially suppressed RB1-deficient TNBC cell proliferation, targeted both the bulk and cancer stem cell fraction, and strongly attenuated xenograft growth. It also cooperated with sulfasalazine, an FDA-approved inhibitor of cystine xCT antiporter, in culture and xenograft assays. Our results suggest that RB1 deficiency promotes cancer cell proliferation in part by enhancing mitochondrial function and identify TIG as a clinically approved drug for RB1-deficient TNBC.


Subject(s)
Gene Expression Regulation, Neoplastic , Mitochondrial Proteins/genetics , Protein Biosynthesis , Retinoblastoma Binding Proteins/deficiency , Triple Negative Breast Neoplasms/metabolism , Ubiquitin-Protein Ligases/deficiency , Animals , Cell Line, Tumor , Female , Gene Amplification , Humans , Mice, Transgenic , Mitochondrial Proteins/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Protein Interaction Maps , Retinoblastoma Binding Proteins/genetics , Transcriptional Activation , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics
13.
Oncotarget ; 7(8): 9060-8, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26814435

ABSTRACT

PTEN loss and PIK3CA activation both promote the accumulation of phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3). While these proteins also have distinct biochemical functions, beyond the regulation of PIP3, little is known about the consequences of these differences in vivo. Here, we directly compared cancer signalling in mammary tumors from MMTV-Cre:Ptenf/f and MMTV-Cre:Pik3ca(LSL-H1047R) mice. Using unsupervised hierarchical clustering we found that whereas MMTV-Cre:Pik3ca(LSL-H1047R)-derived tumors fall into two separate groups, designated squamous-likeEx and class14(Ex), MMTV-Cre:Ptenf/f tumors cluster as one group together with PIK3CA(H1047R) class14(Ex), exhibiting a 'luminal' expression profile. Gene Set Enrichment Analysis (GSEA) of Pten(Δ)ˆ† and PIK3CA(H1047R) class14(Ex) tumors revealed very similar profiles of signalling pathways as well as some interesting differences. Analysis of 18 signalling signatures revealed that PI3K signalling is significantly induced whereas EGFR signalling is significantly reduced in Pten(∆) versus PIK3CA(H1047R) tumors. Thus, Pten(∆) and PIK3CA(H1047R) tumors exhibit discernable differences that may impact tumorigenesis and response to therapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mammary Neoplasms, Experimental/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol Phosphates/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases , ErbB Receptors/metabolism , Mammary Neoplasms, Experimental/classification , Mammary Neoplasms, Experimental/pathology , Mice , Phosphatidylinositol 3-Kinases/metabolism
14.
Cancer Res ; 75(22): 4960-72, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26400062

ABSTRACT

Breast cancer is associated with alterations in a number of growth factor and hormone-regulated signaling pathways. Mouse models of metastatic breast cancer typically feature mutated oncoproteins that activate PI3K, Stat3, and Ras signaling, but the individual and combined roles of these pathways in breast cancer progression are poorly understood. In this study, we examined the relationship between oncogenic pathway activation and breast cancer subtype by analyzing mouse mammary tumor formation in which each pathway was activated singly or pairwise. All three oncogenes showed cooperation during primary tumor formation, but efficient dissemination was only dependent on Ras. In addition, transcriptional profiling demonstrated that Ras induced adenocarcinomas with molecular characteristics related to human basal-like and HER2(+) tumors. In contrast, Ras combined with PIK3CA(H1047R), an oncogenic mutant linked to ERα(+)/luminal breast cancer in humans, induced metastatic luminal B-like tumors. Consistent with these data, elevated Ras signaling was associated with basal-like and HER2(+) subtype tumors in humans and showed a statistically significant negative association with estrogen receptor (ER) signaling across all breast cancer. Despite this, there are luminal tumors with elevated Ras signaling. Importantly, when considered as a continuous variable, Ras pathway activation was strongly linked to reduced survival of patients with ERα(+) disease independent of PI3K or Stat3 activation. Therefore, our studies suggest that Ras activation is a key determinant for dissemination and poor prognosis of ERα(+)/luminal breast cancer in humans, and hormone therapy supplemented with Ras-targeting agents may be beneficial for treating this aggressive subtype.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Invasiveness/pathology , Signal Transduction/physiology , ras Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Disease-Free Survival , Female , Immunohistochemistry , Kaplan-Meier Estimate , Mice , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Oligonucleotide Array Sequence Analysis
15.
Cancer Res ; 75(10): 1936-43, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25808869

ABSTRACT

Claudin-low breast cancer (CLBC) is a poor prognosis disease biologically characterized by stemness and mesenchymal features. These tumors disproportionately affect younger patients and women with African ancestry, causing significant morbidity and mortality, and no effective targeted therapy exists at present. CLBC is thought to originate from mammary stem cells, but little is known on how or why these tumors express a stable epithelial-to-mesenchymal transition phenotype, or what are the driving forces of this disease. Here, we report that Manic Fringe (Mfng), which encodes an O-fucosylpeptide 3-ß-N-acetylglucosaminyltransferase known to modify EGF repeats in the Notch extracellular domain, is highly expressed in CLBC and functions as an oncogene in this context. We show that Mfng modulates Notch activation in human and mouse CLBC cell lines, as well as in mouse mammary gland. Mfng silencing in CLBC cell lines reduced cell migration, tumorsphere formation, and in vivo tumorigenicity associated with a decrease in the stem-like cell population. Mfng deletion in the Lfng(flox/flox);MMTV-Cre mouse model, in which one-third of mammary tumors resemble human CLBC, caused a tumor subtype shift away from CLBC. We identified the phosphoinositide kinase Pik3cg as a direct transcriptional target of Mfng-facilitated RBPJκ-dependent Notch signaling. Indeed, pharmacologic inhibition of PI3Kγ in CLBC cell lines blocked migration and tumorsphere formation. Taken together, our results define Mfng as an oncogene acting through Notch-mediated induction of Pik3cg. Furthermore, they suggest that targeting PI3Kγ may prove beneficial for the treatment of CLBC subtype.


Subject(s)
Breast Neoplasms/enzymology , Class Ib Phosphatidylinositol 3-Kinase/genetics , Claudins/metabolism , Hexosyltransferases/physiology , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Receptors, Notch/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Enzyme Induction , Epithelial-Mesenchymal Transition , Female , Glucosyltransferases , Humans , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Neoplasm Transplantation , Neoplastic Stem Cells/physiology , Phenotype , Signal Transduction
16.
Proc Natl Acad Sci U S A ; 111(52): E5716-23, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25512545

ABSTRACT

Inherited photoreceptor degenerations (IPDs), a group of incurable progressive blinding diseases, are caused by mutations in more than 200 genes, but little is known about the molecular pathogenesis of photoreceptor (PR) death. Increased retinal expression of STAT3 has been observed in response to many retinal insults, including IPDs, but the role of this increase in PR death is unknown. Here, we show that the expression of Stat3 is increased in PRs of the Tg(RHO P347S) and Prph2(rds) (/+) mouse models of IPD and is activated by tyrosine phosphorylation. PR-specific deletion of Stat3 substantially accelerated PR degeneration in both mutant strains. In contrast, increased PR-specific expression of ROSA26 (R26) alleles encoding either WT STAT3 (Stat3(wt)) or the gain-of-function variant STAT3(C) (Stat3(C)) improved PR survival in both models. Moreover, PR signaling in Tg(RHO P347S) mice carrying either a R26-Stat3(wt) or R26-Stat3(C) allele demonstrated increased a-wave amplitude of the scotopic electroretinogram. Phosphorylation of STAT3 at tyrosine 705 was required for the prosurvival effect because an R26-Stat3(Y705F) allele was not protective. The prosurvival role of enhanced Stat3 activity was validated using recombinant adenoassociated virus (rAAV) vector-mediated PR Stat3 expression in Tg(RHO P347S) mice. Our findings (i) establish that the increase in endogenous PR Stat3 expression is a protective response in IPDs, (ii) suggest that therapeutic augmentation of PR Stat3 expression has potential as a common neuroprotective therapy for these disorders, and (iii) indicate that prosurvival molecules whose expression is increased in mutant PRs may have promise as novel therapies for IPDs.


Subject(s)
Genetic Diseases, Inborn/metabolism , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Survival/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Mice , Mice, Transgenic , Photoreceptor Cells, Vertebrate/pathology , RNA, Untranslated/biosynthesis , RNA, Untranslated/genetics , Retinal Degeneration/genetics , Retinal Degeneration/pathology , STAT3 Transcription Factor/genetics
17.
EMBO Mol Med ; 6(12): 1542-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25330770

ABSTRACT

The tumor suppressors Pten and p53 are frequently lost in breast cancer, yet the consequences of their combined inactivation are poorly understood. Here, we show that mammary-specific deletion of Pten via WAP-Cre, which targets alveolar progenitors, induced tumors with shortened latency compared to those induced by MMTV-Cre, which targets basal/luminal progenitors. Combined Pten-p53 mutations accelerated formation of claudin-low, triple-negative-like breast cancer (TNBC) that exhibited hyper-activated AKT signaling and more mesenchymal features relative to Pten or p53 single-mutant tumors. Twenty-four genes that were significantly and differentially expressed between WAP-Cre:Pten/p53 and MMTV-Cre:Pten/p53 tumors predicted poor survival for claudin-low patients. Kinome screens identified eukaryotic elongation factor-2 kinase (eEF2K) inhibitors as more potent than PI3K/AKT/mTOR inhibitors on both mouse and human Pten/p53-deficient TNBC cells. Sensitivity to eEF2K inhibition correlated with AKT pathway activity. eEF2K monotherapy suppressed growth of Pten/p53-deficient TNBC xenografts in vivo and cooperated with doxorubicin to efficiently kill tumor cells in vitro. Our results identify a prognostic signature for claudin-low patients and provide a rationale for using eEF2K inhibitors for treatment of TNBC with elevated AKT signaling.


Subject(s)
Elongation Factor 2 Kinase/metabolism , Gene Deletion , PTEN Phosphohydrolase/genetics , Triple Negative Breast Neoplasms/enzymology , Tumor Suppressor Protein p53/genetics , Animals , Elongation Factor 2 Kinase/antagonists & inhibitors , Elongation Factor 2 Kinase/genetics , Enzyme Inhibitors/pharmacology , Epithelium/enzymology , Epithelium/metabolism , Female , Humans , Mammary Glands, Human/enzymology , Mammary Glands, Human/metabolism , Mice , Mice, Inbred C57BL , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism
18.
Cancer Cell ; 26(4): 452-4, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25314075

ABSTRACT

Human bladder cancers harbor deletions and point mutations in genes coding for Notch receptors and proteins involved in Notch signaling. This leads to elevated MAPK pathway activation, as direct Notch-mediated transcription of MAPK phosphatase DUSP is lost. These bladder tumors, with impaired Notch signaling, also show basal differentiation.


Subject(s)
Receptors, Notch/genetics , Urinary Bladder Neoplasms/pathology , Humans , Point Mutation , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/metabolism
19.
Am J Physiol Endocrinol Metab ; 307(7): E596-610, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25117410

ABSTRACT

The multidrug resistance efflux transporter ATP-binding cassette subfamily G member 2 (ABCG2) is not only overexpressed in certain drug-resistant cancers but is also highly expressed in the mammary gland during lactation, carrying xenobiotics and nutrients into milk. We sought to investigate the molecular mechanisms involved in the upregulation of ABCG2 during lactation. Expression profiling of different mouse Abcg2 mRNA isoforms (E1a, E1b, and E1c) revealed that E1b is predominantly expressed and induced in the lactating mouse mammary gland. Despite this induction, analyses of CpG methylation status and published ChIP-seq datasets reveal that E1b promoter sequences in the virgin gland are already hypomethylated and marked with the open chromatin histone mark H3K4me2. Using a forced-weaning model to shut down lactation, we found that within 24 h there was a significant reduction in Abcg2 mRNA expression and a loss of signal transducer and activator of transcription-5 (STAT5) occupancy at the mouse Abcg2 gene. Luciferase reporter assays further showed that some of these STAT5-binding regions that contained interferon-γ-activated sequence (GAS) motifs function as an enhancer after prolactin treatment. We conclude that Abcg2 is already poised for expression in the virgin mammary gland and that STAT5 plays an important role in Abcg2 expression during lactation.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Epigenesis, Genetic , Epithelial Cells/metabolism , Lactation/genetics , Mammary Glands, Animal/metabolism , RNA Isoforms/genetics , RNA, Messenger/genetics , STAT5 Transcription Factor/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , CpG Islands , DNA Methylation , Female , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Promoter Regions, Genetic , Signal Transduction , Up-Regulation
20.
Neoplasia ; 16(2): 158-67, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24709423

ABSTRACT

Elevated Notch ligand and receptor expression has been associated with aggressive forms of prostate cancer, suggesting a role for Notch signaling in regulation of prostate tumor initiation and progression. Here, we report a critical role for Lunatic Fringe (Lfng), which encodes an O-fucosylpeptide 3-ß-N-acetylglucosaminyltransferase known to modify epidermal growth factor repeats of Notch receptor proteins, in regulation of prostate epithelial differentiation and proliferation, as well as in prostate tumor suppression. Deletion of Lfng in mice caused altered Notch activation in the prostate, associated with elevated accumulation of Notch1, Notch2, and Notch4 intracellular domains, decreased levels of the putative Notch3 intracellular fragment, as well as increased expression of Hes1, Hes5, and Hey2. Loss of Lfng resulted in expansion of the basal layer, increased proliferation of both luminal and basal cells, and ultimately, prostatic intraepithelial neoplasia. The Lfng-null prostate showed down-regulation of prostatic tumor suppressor gene NKX3.1 and increased androgen receptor expression. Interestingly, expression of LFNG and NKX3.1 were positively correlated in publically available human prostate cancer data sets. Knockdown of LFNG in DU-145 prostate cancer cells led to expansion of CD44(+)CD24(-) and CD49f(+)CD24(-) stem/progenitor-like cell population associated with enhanced prostatosphere-forming capacity. Taken together, these data revealed a tumor-suppressive role for Lfng in the prostate through differential regulation of Notch signaling.


Subject(s)
Genes, Tumor Suppressor , Glycosyltransferases/physiology , Prostate/metabolism , Prostatic Neoplasms/genetics , Receptors, Notch/metabolism , Animals , Cell Proliferation , Gene Expression , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Male , Mice, Knockout , Prostate/pathology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Receptor, Notch3 , Receptor, Notch4 , Receptors, Androgen/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...