Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 81(2): 386-397.e7, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33340488

ABSTRACT

In tumors, nutrient availability and metabolism are known to be important modulators of growth signaling. However, it remains elusive whether cancer cells that are growing out in the metastatic niche rely on the same nutrients and metabolic pathways to activate growth signaling as cancer cells within the primary tumor. We discovered that breast-cancer-derived lung metastases, but not the corresponding primary breast tumors, use the serine biosynthesis pathway to support mTORC1 growth signaling. Mechanistically, pyruvate uptake through Mct2 supported mTORC1 signaling by fueling serine biosynthesis-derived α-ketoglutarate production in breast-cancer-derived lung metastases. Consequently, expression of the serine biosynthesis enzyme PHGDH was required for sensitivity to the mTORC1 inhibitor rapamycin in breast-cancer-derived lung tumors, but not in primary breast tumors. In summary, we provide in vivo evidence that the metabolic and nutrient requirements to activate growth signaling differ between the lung metastatic niche and the primary breast cancer site.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Phosphoglycerate Dehydrogenase/genetics , Serine/biosynthesis , Animals , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Humans , Ketoglutaric Acids/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Phosphoglycerate Dehydrogenase/metabolism , Pyruvic Acid/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sirolimus/pharmacology
2.
PLoS Genet ; 20(5): e1011236, 2024 May.
Article in English | MEDLINE | ID: mdl-38722825

ABSTRACT

Patients with ER-negative breast cancer have the worst prognosis of all breast cancer subtypes, often experiencing rapid recurrence or progression to metastatic disease shortly after diagnosis. Given that metastasis is the primary cause of mortality in most solid tumors, understanding metastatic biology is crucial for effective intervention. Using a mouse systems genetics approach, we previously identified 12 genes associated with metastatic susceptibility. Here, we extend those studies to identify Resf1, a poorly characterized gene, as a novel metastasis susceptibility gene in ER- breast cancer. Resf1 is a large, unstructured protein with an evolutionarily conserved intron-exon structure, but with poor amino acid conservation. CRISPR or gene trap mouse models crossed to the Polyoma Middle-T antigen genetically engineered mouse model (MMTV-PyMT) demonstrated that reduction of Resf1 resulted in a significant increase in tumor growth, a shortened overall survival time, and increased incidence and number of lung metastases, consistent with patient data. Furthermore, an analysis of matched tail and primary tissues revealed loss of the wildtype copy in tumor tissue, consistent with Resf1 being a tumor suppressor. Mechanistic analysis revealed a potential role of Resf1 in transcriptional control through association with compound G4 quadruplexes in expressed sequences, particularly those associated with ribosomal biogenesis. These results suggest that loss of Resf1 enhances tumor progression in ER- breast cancer through multiple alterations in both transcriptional and translational control.


Subject(s)
Repressor Proteins , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , Cell Line, Tumor , G-Quadruplexes , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Neoplasm Metastasis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism
3.
PLoS Genet ; 18(6): e1010271, 2022 06.
Article in English | MEDLINE | ID: mdl-35727842

ABSTRACT

The TGF-ß-regulated Chloride Intracellular Channel 4 (CLIC4) is an essential participant in the formation of breast cancer stroma. Here, we used data available from the TCGA and METABRIC datasets to show that CLIC4 expression was higher in breast cancers from younger women and those with early-stage metastatic disease. Elevated CLIC4 predicted poor outcome in breast cancer patients and was linked to the TGF-ß pathway. However, these associations did not reveal the underlying biological contribution of CLIC4 to breast cancer progression. Constitutive ablation of host Clic4 in two murine metastatic breast cancer models nearly eliminated lung metastases without reducing primary tumor weight, while tumor cells ablated of Clic4 retained metastatic capability in wildtype hosts. Thus, CLIC4 was required for host metastatic competence. Pre- and post-metastatic proteomic analysis identified circulating pro-metastatic soluble factors that differed in tumor-bearing CLIC4-deficient and wildtype hosts. Vascular abnormalities and necrosis increased in primary tumors from CLIC4-deficient hosts. Transcriptional profiles of both primary tumors and pre-metastatic lungs of tumor-bearing CLIC4-deficient hosts were consistent with a microenvironment where inflammatory pathways were elevated. Altogether, CLIC4 expression in human breast cancers may serve as a prognostic biomarker; therapeutic targeting of CLIC4 could reduce primary tumor viability and host metastatic competence.


Subject(s)
Breast Neoplasms , Chloride Channels , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chloride Channels/biosynthesis , Chloride Channels/genetics , Female , Humans , Mice , Neoplasm Metastasis , Proteomics , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
4.
PLoS Genet ; 15(5): e1008020, 2019 05.
Article in English | MEDLINE | ID: mdl-31125342

ABSTRACT

Breast cancer is the second leading cause of cancer-related deaths in the United States, with the majority of these deaths due to metastatic lesions rather than the primary tumor. Thus, a better understanding of the etiology of metastatic disease is crucial for improving survival. Using a haplotype mapping strategy in mouse and shRNA-mediated gene knockdown, we identified Rnaseh2c, a scaffolding protein of the heterotrimeric RNase H2 endoribonuclease complex, as a novel metastasis susceptibility factor. We found that the role of Rnaseh2c in metastatic disease is independent of RNase H2 enzymatic activity, and immunophenotyping and RNA-sequencing analysis revealed engagement of the T cell-mediated adaptive immune response. Furthermore, the cGAS-Sting pathway was not activated in the metastatic cancer cells used in this study, suggesting that the mechanism of immune response in breast cancer is different from the mechanism proposed for Aicardi-Goutières Syndrome, a rare interferonopathy caused by RNase H2 mutation. These results suggest an important novel, non-enzymatic role for RNASEH2C during breast cancer progression and add Rnaseh2c to a panel of genes we have identified that together could determine patients with high risk for metastasis. These results also highlight a potential new target for combination with immunotherapies and may contribute to a better understanding of the etiology of Aicardi-Goutières Syndrome autoimmunity.


Subject(s)
Adaptive Immunity , Autoimmune Diseases of the Nervous System/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Nervous System Malformations/genetics , Ribonuclease H/genetics , Animals , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/mortality , Autoimmune Diseases of the Nervous System/pathology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lymphatic Metastasis , Mice , Mice, Nude , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Nervous System Malformations/immunology , Nervous System Malformations/mortality , Nervous System Malformations/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/immunology , Sequence Analysis, RNA , Signal Transduction , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
PLoS Genet ; 12(9): e1006267, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27656887

ABSTRACT

Breast cancer mortality is primarily due to metastasis rather than primary tumors, yet relatively little is understood regarding the etiology of metastatic breast cancer. Previously, using a mouse genetics approach, we demonstrated that inherited germline polymorphisms contribute to metastatic disease, and that these single nucleotide polymorphisms (SNPs) could be used to predict outcome in breast cancer patients. In this study, a backcross between a highly metastatic (FVB/NJ) and low metastatic (MOLF/EiJ) mouse strain identified Arntl2, a gene encoding a circadian rhythm transcription factor, as a metastasis susceptibility gene associated with progression, specifically in estrogen receptor-negative breast cancer patients. Integrated whole genome sequence analysis with DNase hypersensitivity sites reveals SNPs in the predicted promoter of Arntl2. Using CRISPR/Cas9-mediated substitution of the MOLF promoter, we demonstrate that the SNPs regulate Arntl2 transcription and affect metastatic burden. Finally, analysis of SNPs associated with ARNTL2 expression in human breast cancer patients revealed reproducible associations of ARNTL2 expression quantitative trait loci (eQTL) SNPs with disease-free survival, consistent with the mouse studies.

6.
PLoS Genet ; 12(1): e1005820, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26807845

ABSTRACT

Accumulating evidence supports the role of an aberrant transcriptome as a driver of metastatic potential. Deadenylation is a general regulatory node for post-transcriptional control by microRNAs and other determinants of RNA stability. Previously, we demonstrated that the CCR4-NOT scaffold component Cnot2 is an inherited metastasis susceptibility gene. In this study, using orthotopic metastasis assays and genetically engineered mouse models, we show that one of the enzymatic subunits of the CCR4-NOT complex, Cnot7, is also a metastasis modifying gene. We demonstrate that higher expression of Cnot7 drives tumor cell autonomous metastatic potential, which requires its deadenylase activity. Furthermore, metastasis promotion by CNOT7 is dependent on interaction with CNOT1 and TOB1. CNOT7 ribonucleoprotein-immunoprecipitation (RIP) and integrated transcriptome wide analyses reveal that CNOT7-regulated transcripts are enriched for a tripartite 3'UTR motif bound by RNA-binding proteins known to complex with CNOT7, TOB1, and CNOT1. Collectively, our data support a model of CNOT7, TOB1, CNOT1, and RNA-binding proteins collectively exerting post-transcriptional control on a metastasis suppressive transcriptional program to drive tumor cell metastasis.


Subject(s)
Breast Neoplasms/genetics , Mammary Neoplasms, Animal/genetics , Receptors, CCR4/genetics , Transcription Factors/genetics , Transcriptome/genetics , Animals , Breast Neoplasms/pathology , Exoribonucleases , Female , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Metastasis , RNA Stability/genetics , Repressor Proteins
7.
PLoS Genet ; 12(4): e1005989, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27074153

ABSTRACT

Metastasis remains the primary cause of patient morbidity and mortality in solid tumors and is due to the action of a large number of tumor-autonomous and non-autonomous factors. Here we report the results of a genome-wide integrated strategy to identify novel metastasis susceptibility candidate genes and molecular pathways in breast cancer metastasis. This analysis implicates a number of transcriptional regulators and suggests cell-mediated immunity is an important determinant. Moreover, the analysis identified novel or FDA-approved drugs as potentially useful for anti-metastatic therapy. Further explorations implementing this strategy may therefore provide a variety of information for clinical applications in the control and treatment of advanced neoplastic disease.


Subject(s)
Genetic Predisposition to Disease , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/genetics , Animals , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Mammary Neoplasms, Animal/diagnosis , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred NZB , Mice, Transgenic , Nectins , Promyelocytic Leukemia Zinc Finger Protein , RNA Interference , RNA, Small Interfering/genetics , Rosiglitazone , Thiazolidinediones/pharmacology
8.
Genome Res ; 24(2): 227-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24322557

ABSTRACT

Metastasis is the result of stochastic genomic and epigenetic events leading to gene expression profiles that drive tumor dissemination. Here we exploit the principle that metastatic propensity is modified by the genetic background to generate prognostic gene expression signatures that illuminate regulators of metastasis. We also identify multiple microRNAs whose germline variation is causally linked to tumor progression and metastasis. We employ network analysis of global gene expression profiles in tumors derived from a panel of recombinant inbred mice to identify a network of co-expressed genes centered on Cnot2 that predicts metastasis-free survival. Modulating Cnot2 expression changes tumor cell metastatic potential in vivo, supporting a functional role for Cnot2 in metastasis. Small RNA sequencing of the same tumor set revealed a negative correlation between expression of the Mir216/217 cluster and tumor progression. Expression quantitative trait locus analysis (eQTL) identified cis-eQTLs at the Mir216/217 locus, indicating that differences in expression may be inherited. Ectopic expression of Mir216/217 in tumor cells suppressed metastasis in vivo. Finally, small RNA sequencing and mRNA expression profiling data were integrated to reveal that miR-3470a/b target a high proportion of network transcripts. In vivo analysis of Mir3470a/b demonstrated that both promote metastasis. Moreover, Mir3470b is a likely regulator of the Cnot2 network as its overexpression down-regulated expression of network hub genes and enhanced metastasis in vivo, phenocopying Cnot2 knockdown. The resulting data from this strategy identify Cnot2 as a novel regulator of metastasis and demonstrate the power of our systems-level approach in identifying modifiers of metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Neoplasm Metastasis/genetics , Neoplasms/genetics , Repressor Proteins/genetics , Animals , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Mice , MicroRNAs/genetics , Neoplasm Metastasis/pathology , Neoplasms/pathology , Quantitative Trait Loci/genetics , RNA, Messenger/genetics
9.
Proc Natl Acad Sci U S A ; 109(8): 3184-9, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22308418

ABSTRACT

Metastatic disease is the proximal cause of mortality for most cancers and remains a significant problem for the clinical management of neoplastic disease. Recent advances in global transcriptional analysis have enabled better prediction of individuals likely to progress to metastatic disease. However, minimal overlap between predictive signatures has precluded easy identification of key biological processes contributing to the prometastatic transcriptional state. To overcome this limitation, we have applied network analysis to two independent human breast cancer datasets and three different mouse populations developed for quantitative analysis of metastasis. Analysis of these datasets revealed that the gene membership of the networks is highly conserved within and between species, and that these networks predicted distant metastasis free survival. Furthermore these results suggest that susceptibility to metastatic disease is cell-autonomous in estrogen receptor-positive tumors and associated with the mitotic spindle checkpoint. In contrast, nontumor genetics and pathway activities-associated stromal biology are significant modifiers of the rate of metastatic spread of estrogen receptor-negative tumors. These results suggest that the application of network analysis across species may provide a robust method to identify key biological programs associated with human cancer progression.


Subject(s)
Disease Susceptibility , Gene Regulatory Networks/genetics , Neoplasm Metastasis/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Databases, Genetic , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Estrogen/metabolism , Species Specificity , Tetraspanin 25/metabolism
10.
PLoS Genet ; 8(5): e1002735, 2012 May.
Article in English | MEDLINE | ID: mdl-22693453

ABSTRACT

Accumulating evidence suggests that breast cancer metastatic progression is modified by germline polymorphism, although specific modifier genes have remained largely undefined. In the current study, we employ the MMTV-PyMT transgenic mouse model and the AKXD panel of recombinant inbred mice to identify AT-rich interactive domain 4B (Arid4b; NM_194262) as a breast cancer progression modifier gene. Ectopic expression of Arid4b promoted primary tumor growth in vivo as well as increased migration and invasion in vitro, and the phenotype was associated with polymorphisms identified between the AKR/J and DBA/2J alleles as predicted by our genetic analyses. Stable shRNA-mediated knockdown of Arid4b caused a significant reduction in pulmonary metastases, validating a role for Arid4b as a metastasis modifier gene. ARID4B physically interacts with the breast cancer metastasis suppressor BRMS1, and we detected differential binding of the Arid4b alleles to histone deacetylase complex members mSIN3A and mSDS3, suggesting that the mechanism of Arid4b action likely involves interactions with chromatin modifying complexes. Downregulation of the conserved Tpx2 gene network, which is comprised of many factors regulating cell cycle and mitotic spindle biology, was observed concomitant with loss of metastatic efficiency in Arid4b knockdown cells. Consistent with our genetic analysis and in vivo experiments in our mouse model system, ARID4B expression was also an independent predictor of distant metastasis-free survival in breast cancer patients with ER+ tumors. These studies support a causative role of ARID4B in metastatic progression of breast cancer.


Subject(s)
Cell Movement/genetics , DNA-Binding Proteins/genetics , Mammary Neoplasms, Animal/genetics , Repressor Proteins/genetics , Alleles , Animals , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Microtubule-Associated Proteins/metabolism , Neoplasm Metastasis , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics , Sin3 Histone Deacetylase and Corepressor Complex
11.
PLoS Genet ; 8(9): e1002926, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028344

ABSTRACT

Metastasis is a complex process utilizing both tumor-cell-autonomous properties and host-derived factors, including cellular immunity. We have previously shown that germline polymorphisms can modify tumor cell metastatic capabilities through cell-autonomous mechanisms. However, how metastasis susceptibility genes interact with the tumor stroma is incompletely understood. Here, we employ a complex genetic screen to identify Cadm1 as a novel modifier of metastasis. We demonstrate that Cadm1 can specifically suppress metastasis without affecting primary tumor growth. Unexpectedly, Cadm1 did not alter tumor-cell-autonomous properties such as proliferation or invasion, but required the host's adaptive immune system to affect metastasis. The metastasis-suppressing effect of Cadm1 was lost in mice lacking T cell-mediated immunity, which was partially phenocopied by depleting CD8(+) T cells in immune-competent mice. Our data show a novel function for Cadm1 in suppressing metastasis by sensitizing tumor cells to immune surveillance mechanisms, and this is the first report of a heritable metastasis susceptibility gene engaging tumor non-autonomous factors.


Subject(s)
Cell Adhesion Molecules/genetics , Genes, Tumor Suppressor , Immunity, Cellular/genetics , Immunoglobulins/genetics , Neoplasm Metastasis/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Immunoglobulins/metabolism , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/immunology , Prognosis
12.
Nat Genet ; 37(10): 1055-62, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142231

ABSTRACT

We previously identified loci in the mouse genome that substantially influence the metastatic efficiency of mammary tumors. Here, we present data supporting the idea that the signal transduction molecule, Sipa1, is a candidate for underlying the metastasis efficiency modifier locus Mtes1. Analysis of candidate genes identified a nonsynonymous amino acid polymorphism in Sipa1 that affects the Sipa1 Rap-GAP function. Spontaneous metastasis assays using cells ectopically expressing Sipa1 or cells with knocked-down Sipa1 expression showed that metastatic capacity was correlated with cellular Sipa1 levels. We examined human expression data and found that they were consistent with the idea that Sipa1 concentration has a role in metastasis. Taken together, these data suggest that the Sipa1 polymorphism is one of the genetic polymorphisms underlying the Mtes1 locus. This report is also the first demonstration, to our knowledge, of a constitutional genetic polymorphism affecting tumor metastasis.


Subject(s)
GTPase-Activating Proteins/genetics , Neoplasm Metastasis/genetics , Nuclear Proteins/genetics , Polymorphism, Genetic , rap GTP-Binding Proteins/genetics , Animals , Biological Assay , COS Cells , Cell Adhesion/genetics , Chlorocebus aethiops , GTPase-Activating Proteins/metabolism , Humans , Mice , Mutation , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , Transcriptional Activation , rap GTP-Binding Proteins/metabolism
13.
bioRxiv ; 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38410432

ABSTRACT

Acetylation of protein and RNA represent a critical event for development and cancer progression. NAT10 is the only known RNA acetylase that catalyzes the N4-actylcytidine (ac4C) modification of RNAs. Here, we show that the loss of NAT10 significantly decreases lung metastasis in allograft and genetically engineered mouse models of breast cancer. NAT10 interacts with a mechanosensitive, metastasis susceptibility protein complex at the nuclear pore. In addition to its canonical role in RNA acetylation, we find that NAT10 interacts with p300 at gene enhancers. NAT10 loss is associated with p300 mislocalization into heterochromatin regions. NAT10 depletion disrupts enhancer organization, leading to alteration of gene transcription necessary for metastatic progression, including reduced myeloid cell-recruiting chemokines that results in a less metastasis-prone tumor microenvironment. Our study uncovers a distinct role of NAT10 in enhancer organization of metastatic tumor cells and suggests its involvement in the tumor-immune crosstalk dictating metastatic outcomes.

14.
Gastroenterology ; 142(4): 957-966.e12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22202459

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is an aggressive malignancy; its mechanisms of development and progression are poorly understood. We used an integrative approach to identify HCC driver genes, defined as genes whose copy numbers associate with gene expression and cancer progression. METHODS: We combined data from high-resolution, array-based comparative genomic hybridization and transcriptome analysis of HCC samples from 76 patients with hepatitis B virus infection with data on patient survival times. Candidate genes were functionally validated using in vitro and in vivo models. RESULTS: Unsupervised analyses of array comparative genomic hybridization data associated loss of chromosome 8p with poor outcome (reduced survival time); somatic copy number alterations correlated with expression of 27.3% of genes analyzed. We associated expression levels of 10 of these genes with patient survival times in 2 independent cohorts (comprising 319 cases of HCC with mixed etiology) and 3 breast cancer cohorts (637 cases). Among the 10-gene signature, a cluster of 6 genes on 8p, (DLC1, CCDC25, ELP3, PROSC, SH2D4A, and SORBS3) were deleted in HCCs from patients with poor outcomes. In vitro and in vivo analyses indicated that the products of PROSC, SH2D4A, and SORBS3 have tumor-suppressive activities, along with the known tumor suppressor gene DLC1. CONCLUSIONS: We used an unbiased approach to identify 10 genes associated with HCC progression. These might be used in assisting diagnosis and to stage tumors based on gene expression patterns.


Subject(s)
Carcinoma, Hepatocellular/genetics , Chromosome Deletion , Chromosomes, Human, Pair 8 , Liver Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , China , Comparative Genomic Hybridization , Disease Progression , Female , GTPase-Activating Proteins/genetics , Gene Dosage , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Histone Acetyltransferases/genetics , Humans , Intracellular Signaling Peptides and Proteins , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Middle Aged , Minnesota , Multivariate Analysis , Muscle Proteins , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Prognosis , Proteins/genetics , Proteins/metabolism , Reproducibility of Results , Risk Assessment , Risk Factors , Time Factors , Transfection , Tumor Burden , Tumor Suppressor Proteins/genetics
15.
Cancer Res ; 83(8): 1280-1298, 2023 04 14.
Article in English | MEDLINE | ID: mdl-36799863

ABSTRACT

Understanding functional interactions between cancer mutations is an attractive strategy for discovering unappreciated cancer pathways and developing new combination therapies to improve personalized treatment. However, distinguishing driver gene pairs from passenger pairs remains challenging. Here, we designed an integrated omics approach to identify driver gene pairs by leveraging genetic interaction analyses of top mutated breast cancer genes and the proteomics interactome data of their encoded proteins. This approach identified that PIK3CA oncogenic gain-of-function (GOF) and CBFB loss-of-function (LOF) mutations cooperate to promote breast tumor progression in both mice and humans. The transcription factor CBFB localized to mitochondria and moonlighted in translating the mitochondrial genome. Mechanistically, CBFB enhanced the binding of mitochondrial mRNAs to TUFM, a mitochondrial translation elongation factor. Independent of mutant PI3K, mitochondrial translation defects caused by CBFB LOF led to multiple metabolic reprogramming events, including defective oxidative phosphorylation, the Warburg effect, and autophagy/mitophagy addiction. Furthermore, autophagy and PI3K inhibitors synergistically killed breast cancer cells and impaired the growth of breast tumors, including patient-derived xenografts carrying CBFB LOF and PIK3CA GOF mutations. Thus, our study offers mechanistic insights into the functional interaction between mutant PI3K and mitochondrial translation dysregulation in breast cancer progression and provides a strong preclinical rationale for combining autophagy and PI3K inhibitors in precision medicine for breast cancer. SIGNIFICANCE: CBFB-regulated mitochondrial translation is a regulatory step in breast cancer metabolism and synergizes with mutant PI3K in breast cancer progression.


Subject(s)
Breast Neoplasms , Class I Phosphatidylinositol 3-Kinases , Core Binding Factor beta Subunit , Animals , Female , Humans , Mice , Breast Neoplasms/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/pharmacology , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Signal Transduction/genetics
16.
Proc Natl Acad Sci U S A ; 105(17): 6380-5, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18427120

ABSTRACT

Previous work identified the Rap1 GTPase-activating protein Sipa1 as a germ-line-encoded metastasis modifier. The bromodomain protein Brd4 physically interacts with and modulates the enzymatic activity of Sipa1. In vitro analysis of a highly metastatic mouse mammary tumor cell line ectopically expressing Brd4 demonstrates significant reduction of invasiveness without altering intrinsic growth rate. However, a dramatic reduction of tumor growth and pulmonary metastasis was observed after s.c. implantation into mice, implying that activation of Brd4 may somehow be manipulating response to tumor microenvironment in the in vivo setting. Further in vitro analysis shows that Brd4 modulates extracellular matrix gene expression, a class of genes frequently present in metastasis-predictive gene signatures. Microarray analysis of the mammary tumor cell lines identified a Brd4 activation signature that robustly predicted progression and/or survival in multiple human breast cancer datasets analyzed on different microarray platforms. Intriguingly, the Brd4 signature also almost perfectly matches a molecular classifier of low-grade tumors. Taken together, these data suggest that dysregulation of Brd4-associated pathways may play an important role in breast cancer progression and underlies multiple common prognostic signatures.


Subject(s)
Breast Neoplasms/pathology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Databases, Genetic , Extracellular Matrix/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/pathology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Receptors, Estrogen/metabolism , Survival Analysis
17.
Nat Commun ; 12(1): 7216, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903738

ABSTRACT

Mechanical signals from the extracellular microenvironment have been implicated in tumor and metastatic progression. Here, we identify nucleoporin NUP210 as a metastasis susceptibility gene for human estrogen receptor positive (ER+) breast cancer and a cellular mechanosensor. Nup210 depletion suppresses lung metastasis in mouse models of breast cancer. Mechanistically, NUP210 interacts with LINC complex protein SUN2 which connects the nucleus to the cytoskeleton. In addition, the NUP210/SUN2 complex interacts with chromatin via the short isoform of BRD4 and histone H3.1/H3.2 at the nuclear periphery. In Nup210 knockout cells, mechanosensitive genes accumulate H3K27me3 heterochromatin modification, mediated by the polycomb repressive complex 2 and differentially reposition within the nucleus. Transcriptional repression in Nup210 knockout cells results in defective mechanotransduction and focal adhesion necessary for their metastatic capacity. Our study provides an important role of nuclear pore protein in cellular mechanosensation and metastasis.


Subject(s)
Breast Neoplasms/pathology , Heterochromatin/metabolism , Mechanotransduction, Cellular/genetics , Nuclear Pore Complex Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CCCTC-Binding Factor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cytoskeleton/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Focal Adhesions/genetics , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Methyltransferases/metabolism , Mice , Neoplasm Metastasis , Neoplastic Cells, Circulating/metabolism , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Proteins/metabolism , Polymorphism, Genetic , Prognosis , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Microenvironment
18.
J Immunother Cancer ; 9(5)2021 05.
Article in English | MEDLINE | ID: mdl-33958486

ABSTRACT

BACKGROUND: Clinical studies have linked usage of progestins (synthetic progesterone [P4]) to breast cancer risk. However, little is understood regarding the role of native P4, signaling through the progesterone receptor (PR), in breast tumor formation. Recently, we reported a link between PR and immune signaling pathways, showing that P4/PR can repress type I interferon signaling pathways. Given these findings, we sought to investigate whether P4/PR drive immunomodulation in the mammary gland and promote tumor formation. METHODS: To determine the effect of P4 on immune cell populations in the murine mammary gland, mice were treated with P4 or placebo pellets for 21 days. Immune cell populations in the mammary gland, spleen, and inguinal lymph nodes were subsequently analyzed by flow cytometry. To assess the effect of PR overexpression on mammary gland tumor development as well as immune cell populations in the mammary gland, a transgenic mouse model was used in which PR was overexpressed throughout the entire mouse. Immune cell populations were assessed in the mammary glands, spleens, and inguinal lymph nodes of 6-month-old transgenic and control mice by flow cytometry. Transgenic mice were also monitored for mammary gland tumor development over a 2-year time span. Following development of mammary gland tumors, immune cell populations in the tumors and spleens of transgenic and control mice were analyzed by flow cytometry. RESULTS: We found that mice treated with P4 exhibited changes in the mammary gland indicative of an inhibited immune response compared with placebo-treated mice. Furthermore, transgenic mice with PR overexpression demonstrated decreased numbers of immune cell populations in their mammary glands, lymph nodes, and spleens. On long-term monitoring, we determined that multiparous PR-overexpressing mice developed significantly more mammary gland tumors than control mice. Additionally, tumors from PR-overexpressing mice contained fewer infiltrating immune cells. Finally, RNA sequencing analysis of tumor samples revealed that immune-related gene signatures were lower in tumors from PR-overexpressing mice as compared with control mice. CONCLUSION: Together, these findings offer a novel mechanism of P4-driven mammary gland tumor development and provide rationale in investigating the usage of antiprogestin therapies to promote immune-mediated elimination of mammary gland tumors.


Subject(s)
Breast Neoplasms/chemically induced , Cell Transformation, Neoplastic/chemically induced , Mammary Glands, Animal/drug effects , Progesterone/administration & dosage , Receptors, Progesterone/agonists , Tumor Escape/drug effects , Tumor Microenvironment/immunology , Adaptive Immunity/drug effects , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drug Implants , Female , Galectin 4/genetics , Galectin 4/metabolism , Immunity, Innate/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice, Transgenic , Ovariectomy , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction , Time Factors , Tumor Burden/drug effects
19.
J Biol Chem ; 284(42): 28660-73, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19710015

ABSTRACT

There is accumulating evidence for a role of germ line variation in breast cancer metastasis. We have recently identified a novel metastasis susceptibility gene, Rrp1b (ribosomal RNA processing 1 homolog B). Overexpression of Rrp1b in a mouse mammary tumor cell line induces a gene expression signature that predicts survival in breast cancer. Here we extend the analysis of RRP1B function by demonstrating that the Rrp1b activation gene expression signature accurately predicted the outcome in three of four publicly available breast carcinoma gene expression data sets. In addition, we provide insights into the mechanism of RRP1B. Tandem affinity purification demonstrated that RRP1B physically interacts with many nucleosome binding factors, including histone H1X, poly(ADP-ribose) polymerase 1, TRIM28 (tripartite motif-containing 28), and CSDA (cold shock domain protein A). Co-immunofluorescence and co-immunoprecipitation confirmed these interactions and also interactions with heterochromatin protein-1alpha and acetyl-histone H4 lysine 5. Finally, we investigated the effects of ectopic expression of an RRP1B allelic variant previously associated with improved survival in breast cancer. Gene expression analyses demonstrate that, compared with ectopic expression of wild type RRP1B in HeLa cells, the variant RRP1B differentially modulates various transcription factors controlled by TRIM28 and CSDA. These data suggest that RRP1B, a tumor progression and metastasis susceptibility candidate gene, is potentially a dynamic modulator of transcription and chromatin structure.


Subject(s)
Chromatin/chemistry , Chromosomal Proteins, Non-Histone/physiology , Gene Expression Regulation, Neoplastic , Nuclear Proteins/physiology , Amino Acid Motifs , Animals , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Profiling , HeLa Cells , Humans , Mice , Mitosis , Neoplasm Metastasis , Nuclear Proteins/metabolism , Polymorphism, Genetic , RNA, Ribosomal/metabolism , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28
20.
PLoS Genet ; 3(11): e214, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18081427

ABSTRACT

A novel candidate metastasis modifier, ribosomal RNA processing 1 homolog B (Rrp1b), was identified through two independent approaches. First, yeast two-hybrid, immunoprecipitation, and functional assays demonstrated a physical and functional interaction between Rrp1b and the previous identified metastasis modifier Sipa1. In parallel, using mouse and human metastasis gene expression data it was observed that extracellular matrix (ECM) genes are common components of metastasis predictive signatures, suggesting that ECM genes are either important markers or causal factors in metastasis. To investigate the relationship between ECM genes and poor prognosis in breast cancer, expression quantitative trait locus analysis of polyoma middle-T transgene-induced mammary tumor was performed. ECM gene expression was found to be consistently associated with Rrp1b expression. In vitro expression of Rrp1b significantly altered ECM gene expression, tumor growth, and dissemination in metastasis assays. Furthermore, a gene signature induced by ectopic expression of Rrp1b in tumor cells predicted survival in a human breast cancer gene expression dataset. Finally, constitutional polymorphism within RRP1B was found to be significantly associated with tumor progression in two independent breast cancer cohorts. These data suggest that RRP1B may be a novel susceptibility gene for breast cancer progression and metastasis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chromosomal Proteins, Non-Histone/genetics , Nuclear Proteins/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Baltimore , Chromosomal Proteins, Non-Histone/metabolism , Cohort Studies , Disease Progression , Disease Susceptibility , Extracellular Matrix/genetics , Female , GTPase-Activating Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Haplotypes , Humans , Mice , Mice, Inbred Strains , Mutant Proteins/metabolism , Neoplasm Metastasis , Nuclear Proteins/metabolism , Polymorphism, Genetic , Promoter Regions, Genetic , Protein Binding , Quantitative Trait Loci , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL