Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
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
2.
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
3.
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
4.
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
5.
Genome Biol ; 14(11): R125, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24220145

ABSTRACT

BACKGROUND: Human breast cancer is a heterogeneous disease consisting of multiple molecular subtypes. Genetically engineered mouse models are a useful resource for studying mammary cancers in vivo under genetically controlled and immune competent conditions. Identifying murine models with conserved human tumor features will facilitate etiology determinations, highlight the effects of mutations on pathway activation, and should improve preclinical drug testing. RESULTS: Transcriptomic profiles of 27 murine models of mammary carcinoma and normal mammary tissue were determined using gene expression microarrays. Hierarchical clustering analysis identified 17 distinct murine subtypes. Cross-species analyses using three independent human breast cancer datasets identified eight murine classes that resemble specific human breast cancer subtypes. Multiple models were associated with human basal-like tumors including TgC3(1)-Tag, TgWAP-Myc and Trp53-/-. Interestingly, the TgWAPCre-Etv6 model mimicked the HER2-enriched subtype, a group of human tumors without a murine counterpart in previous comparative studies. Gene signature analysis identified hundreds of commonly expressed pathway signatures between linked mouse and human subtypes, highlighting potentially common genetic drivers of tumorigenesis. CONCLUSIONS: This study of murine models of breast carcinoma encompasses the largest comprehensive genomic dataset to date to identify human-to-mouse disease subtype counterparts. Our approach illustrates the value of comparisons between species to identify murine models that faithfully mimic the human condition and indicates that multiple genetically engineered mouse models are needed to represent the diversity of human breast cancers. The reported trans-species associations should guide model selection during preclinical study design to ensure appropriate representatives of human disease subtypes are used.


Subject(s)
Breast Neoplasms/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Transcriptome/genetics , Animals , Breast Neoplasms/classification , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cluster Analysis , Female , Humans , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA
6.
Cancer Cell ; 21(5): 626-641, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22624713

ABSTRACT

Basal-like breast cancers (BLBC) express a luminal progenitor gene signature. Notch receptor signaling promotes luminal cell fate specification in the mammary gland, while suppressing stem cell self-renewal. Here we show that deletion of Lfng, a sugar transferase that prevents Notch activation by Jagged ligands, enhances stem/progenitor cell proliferation. Mammary-specific deletion of Lfng induces basal-like and claudin-low tumors with accumulation of Notch intracellular domain fragments, increased expression of proliferation-associated Notch targets, amplification of the Met/Caveolin locus, and elevated Met and Igf-1R signaling. Human BL breast tumors, commonly associated with JAGGED expression, elevated MET signaling, and CAVEOLIN accumulation, express low levels of LFNG. Thus, reduced LFNG expression facilitates JAG/NOTCH luminal progenitor signaling and cooperates with MET/CAVEOLIN basal-type signaling to promote BLBC.


Subject(s)
Breast Neoplasms/enzymology , Caveolins/metabolism , Cell Transformation, Neoplastic/metabolism , Glycosyltransferases/metabolism , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Experimental/enzymology , Neoplastic Stem Cells/enzymology , Proto-Oncogene Proteins c-met/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium-Binding Proteins/metabolism , Caveolins/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Claudins/metabolism , Databases, Genetic , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Glycosyltransferases/deficiency , Glycosyltransferases/genetics , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Glands, Animal/transplantation , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Middle Aged , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-met/genetics , Receptor, IGF Type 1/metabolism , Receptors, Notch/metabolism , Serrate-Jagged Proteins , Signal Transduction
7.
Oncotarget ; 2(6): 435-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21646685

ABSTRACT

Most human breast tumors have mutations that elevate signaling through a key metabolic pathway that is induced by insulin and a number of growth factors. This pathway serves to activate an enzyme known as phosphatidylinositol 3' kinase (PI3K) as well as to regulate proteins that signal in response to lipid products of PI3K. The specific mutations that activate this pathway in breast cancer can occur in genes coding for tyrosine kinase receptors, adaptor proteins linked to PI3K, catalytic and regulatory subunits of PI3K, serine/threonine kinases that function downstream of PI3K, and also phosphatidylinositol 3' phosphatase tumor suppressors that function to antagonize this pathway. While each genetic change results in net elevation of PI3K pathway signaling, and all major breast cancer subtypes show pathway activation, the specific mutation(s) involved in any one tumor may play an important role in defining tumor subtype, prognosis and even sensitivity to therapy. Here, we describe mouse models of breast cancer with elevated PI3K signaling, and how they may be used to guide development of novel therapeutics.


Subject(s)
Breast Neoplasms/classification , Carcinoma/classification , Phosphatidylinositol 3-Kinases/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/pathology , Female , Humans , Mice , Models, Biological , Signal Transduction/physiology , Tumor Cells, Cultured , Up-Regulation/physiology
8.
Cancer Res ; 71(7): 2706-17, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21324922

ABSTRACT

PIK3CA, which codes for the p110α catalytic subunit of phosphatidylinositol 3-kinase, is one of the most frequently mutated genes in human breast cancer. Here, we describe a mouse model for PIK3CA-induced breast cancer by using the ROSA26 (R26) knock-in system, in which targeted Pik3ca alleles can be activated through transgenic expression of Cre recombinase. We mated Pik3ca(H1047R) and Pik3ca(wt) knock-in lines with MMTV-Cre transgenics, which express Cre in mammary epithelium. Starting at approximately 5 months of age, female R26-Pik3ca(H1047R);MMTV-Cre mice, but not control R26-Pik3ca(wt);MMTV-Cre mice, developed mammary tumors, as well as lymphoid and skin malignancies. R26-Pik3ca(H1047R);MMTV-Cre mammary tumors were typically either adenosquamous carcinoma or adenomyoepithelioma. As p53 is the most commonly mutated gene in breast cancer, we tested for genetic interaction between Pik3ca(H1047R) and p53 loss-of-function mutations in R26-Pik3ca(H1047R);p53(loxP/+);MMTV-Cre mice. This led to decreased survival of double-mutant animals, which developed lymphoma and mammary tumors with rapid kinetics. Mammary tumors that formed in p53(loxP/+);MMTV-Cre conditional mutants were either poorly differentiated adenocarcinoma or spindle cell/EMT, whereas R26-Pik3ca(H1047R);p53(loxP/+);MMTV-Cre mammary tumors were mostly adenosquamous carcinoma or spindle cell/EMT indicating that double-mutant mice develop a distinct spectrum of mammary tumors. Thus, an oncogenic variant of PIK3CA implicated in multiple human breast cancer subtypes can induce a very diverse spectrum of mammary tumors in mice. Furthermore, Pik3ca(H1047R) shows cooperation with p53, which altered the specific tumors that formed. Thus, the two most frequently mutated genes in human breast cancer show cooperation in mammary tumor formation.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Mutation , Phosphatidylinositol 3-Kinases/genetics , Tumor Suppressor Protein p53/genetics , Alleles , Animals , Class I Phosphatidylinositol 3-Kinases , Female , Gene Knock-In Techniques , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Phosphatidylinositol 3-Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/deficiency
9.
Pituitary ; 9(2): 93-9, 2006.
Article in English | MEDLINE | ID: mdl-16832585

ABSTRACT

Disorders of water metabolism are a common complication of pituitary surgery. The primary purpose of this study was to determine the incidence and duration of post-surgical diabetes insipidus (DI) at our institution. Secondary objectives included characterizing the incidence of post-operative hyponatremia as well as delineating factors associated with the onset of these complications. Records of 319 patients who underwent transsphenoidal pituitary surgery at the authors' institution between 1998 and 2005 were reviewed for the presence of disorders of water metabolism using pre-specified criteria. DI was diagnosed in 59 (18.5%) of our patients at a mean time of 13.6 h following surgery. DI resolved in nearly half of our patients within one week. Approximately 80% of our patients enjoyed resolution of DI at a mean time of 2.9 months following surgery. Duration of DI was not significantly influenced by tumor size or location. Additionally, 28 (8.8%) of our patients exhibited a period of hyponatremia at a mean time of 4 days following surgery. One quarter of these patients carried a diagnosis of Cushing's disease. We herein report an incidence of DI as well as hyponatremia within our post-operative population comparable to that reported by other high-volume pituitary centers. Over half of our patients still exhibited DI at the time of discharge, therefore, patient education regarding the treatment of DI, signs of its resolution, and symptoms consistent with the onset of hyponatremia should be an integral part of every hospitalization.


Subject(s)
Diabetes Insipidus, Neurogenic/epidemiology , Hyponatremia/epidemiology , Pituitary Diseases/surgery , Adult , Diabetes Insipidus, Neurogenic/etiology , Disease Progression , Female , Humans , Hyponatremia/etiology , Incidence , Male , Middle Aged , Retrospective Studies , Risk Factors , Tennessee
SELECTION OF CITATIONS
SEARCH DETAIL
...