Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Sci ; 111(7): 2336-2348, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32437590

ABSTRACT

Dietary fat consumption during accelerated stages of mammary gland development, such as peripubertal maturation or pregnancy, is known to increase the risk for breast cancer. However, the underlying molecular mechanisms are not fully understood. Here we examined the gene expression profile of mouse mammary epithelial cells (MMECs) on exposure to a high-fat diet (HFD) or control diet (CD). Trp53-/- female mice were fed with the experimental diets for 5 weeks during the peripubertal period (3-8 weeks of age). The treatment showed no significant difference in body weight between the HFD-fed mice and CD-fed mice. However, gene set enrichment analysis predicted a significant enrichment of c-Myc target genes in animals fed HFD. Furthermore, we detected enhanced activity and stabilization of c-Myc protein in MMECs exposed to a HFD. This was accompanied by augmented c-Myc phosphorylation at S62 with a concomitant increase in ERK phosphorylation. Moreover, MMECs derived from HFD-fed Trp53-/- mouse showed increased colony- and sphere-forming potential that was dependent on c-Myc. Further, oleic acid, a major fatty acid constituent of the HFD, and TAK-875, an agonist to G protein-coupled receptor 40 (a receptor for oleic acid), enhanced c-Myc stabilization and MMEC proliferation. Overall, our data indicate that HFD influences MMECs by stabilizing an oncoprotein, pointing to a novel mechanism underlying dietary fat-mediated mammary carcinogenesis.


Subject(s)
Diet, High-Fat , Epithelium/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Proto-Oncogene Proteins c-myc/metabolism , Sexual Maturation , Animals , Cell Line, Tumor , Female , Genes, p53 , Humans , Mice , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Proto-Oncogene Proteins c-myc/genetics , Puberty , Tumor Cells, Cultured
2.
Cancer Res ; 79(15): 3903-3915, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31189648

ABSTRACT

Cancer cell-intrinsic properties caused by oncogenic mutations have been well characterized; however, how specific oncogenes and tumor suppressors impact the tumor microenvironment (TME) is not well understood. Here, we present a novel non-cell-autonomous function of the retinoblastoma (RB) tumor suppressor in controlling the TME. RB inactivation stimulated tumor growth and neoangiogenesis in a syngeneic and orthotropic murine soft-tissue sarcoma model, which was associated with recruitment of tumor-associated macrophages (TAM) and immunosuppressive cells such as Gr1+CD11b+ myeloid-derived suppressor cells (MDSC) or Foxp3+ regulatory T cells (Treg). Gene expression profiling and analysis of genetically engineered mouse models revealed that RB inactivation increased secretion of the chemoattractant CCL2. Furthermore, activation of the CCL2-CCR2 axis in the TME promoted tumor angiogenesis and recruitment of TAMs and MDSCs into the TME in several tumor types including sarcoma and breast cancer. Loss of RB increased fatty acid oxidation (FAO) by activating AMP-activated protein kinase that led to inactivation of acetyl-CoA carboxylase, which suppresses FAO. This promoted mitochondrial superoxide production and JNK activation, which enhanced CCL2 expression. These findings indicate that the CCL2-CCR2 axis could be an effective therapeutic target in RB-deficient tumors. SIGNIFICANCE: These findings demonstrate the cell-nonautonomous role of the tumor suppressor retinoblastoma in the tumor microenvironment, linking retinoblastoma loss to immunosuppression.


Subject(s)
Chemokine CCL2/metabolism , Retinoblastoma Protein/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Chemokine CCL2/biosynthesis , Female , Gene Expression Profiling/methods , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/metabolism , Retinoblastoma Protein/deficiency , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology , Tumor Microenvironment , Up-Regulation
3.
Oncotarget ; 8(8): 13872-13885, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28099924

ABSTRACT

We established an in vitro cell culture system to determine novel activities of the retinoblastoma (Rb) protein during tumor progression. Rb depletion in p53-null mouse-derived soft tissue sarcoma cells induced a spherogenic phenotype. Cells retrieved from Rb-depleted spheres exhibited slower proliferation and less efficient BrdU incorporation, however, much higher spherogenic activity and aggressive behavior. We discovered six miRNAs, including mmu-miR-18a, -25, -29b, -140, -337, and -1839, whose expression levels correlated tightly with the Rb status and spherogenic activity. Among these, mmu-miR-140 appeared to be positively controlled by Rb and to antagonize the effect of Rb depletion on spherogenesis and tumorigenesis. Furthermore, among genes potentially targeted by mmu-miR-140, Il-6 was upregulated by Rb depletion and downregulated by mmu-mir-140 overexpression. Altogether, we demonstrate the possibility that mmu-mir-140 mediates the Rb function to downregulate Il-6 by targeting its 3'-untranslated region. Finally, we detected the same relationship among RB, hsa-miR-140 and IL-6 in a human breast cancer cell line MCF-7. Because IL-6 is a critical modulator of malignant features of cancer cells and the RB pathway is impaired in the majority of cancers, hsa-miR-140 might be a promising therapeutic tool that disrupts linkage between tumor suppressor inactivation and pro-inflammatory cytokine response.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Interleukin-6/metabolism , MicroRNAs/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Retinoblastoma Protein/metabolism , Animals , Cell Line, Tumor , Humans , Immunoblotting , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Retinoblastoma Protein/genetics , Transcriptome
4.
Mol Carcinog ; 55(12): 1974-1989, 2016 12.
Article in English | MEDLINE | ID: mdl-26621780

ABSTRACT

Mutations in RB and PTEN are linked to castration resistance and poor prognosis in prostate cancer. Identification of genes that are regulated by these tumor suppressors in a context that recapitulates cancer progression may be beneficial for discovering novel therapeutic targets. Although various genetically engineered mice thus far provided tumor models with various pathological stages, they are not ideal for detecting dynamic changes in gene transcription. Additionally, it is difficult to achieve an effect specific to tumor progression via gain of functions of these genes. In this study, we developed an in vitro model to help identify RB- and PTEN-loss signatures during the malignant progression of prostate cancers. Trp53-/- ; Rbf/f , Trp53-/- ; Ptenf/f , and Trp53-/- ; Rbf/f ; Ptenf/f prostate epithelial cells were infected with AD-LacZ or AD-Cre. We found that deletion of Rb, Pten or both stimulated prostasphere formation and tumor development in immune-compromised mice. The GO analysis of genes affected by the deletion of Rb or Pten in Trp53-/- prostate epithelial cells identified a number of genes encoding cytokines, chemokines and extracellular matrix remodeling factors, but only few genes related to cell cycle progression. Two genes (Il-6 and Lox) were further analyzed. Blockade of Il-6 signaling and depletion of Lox significantly attenuated prostasphere formation in 3D culture, and in the case of IL-6, strongly suppressed tumor growth in vivo. These findings suggest that our in vitro model may be instrumental in identifying novel therapeutic targets of prostate cancer progression, and further underscore IL-6 and LOX as promising therapeutic targets. © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinogenesis/pathology , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/genetics , Prostate/pathology , Prostatic Neoplasms/pathology , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/genetics , Cells, Cultured , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Deletion , Male , Mice , Mice, Knockout , Prostate/metabolism , Prostatic Neoplasms/genetics , Signal Transduction
5.
Stem Cells ; 33(5): 1657-69, 2015 May.
Article in English | MEDLINE | ID: mdl-25694388

ABSTRACT

Retinoblastoma tumor suppressor protein (RB) is inactivated more frequently during tumor progression than during tumor initiation. However, its exact role in controlling the malignant features associated with tumor progression is poorly understood. We established in vivo and in vitro models to investigate the undifferentiated state induced by Rb inactivation. Rb heterozygous mice develop well-differentiated thyroid medullary carcinoma. We found that additional deletion of Trp53, without change in lineage, converted these Rb-deficient tumors to a poorly differentiated type associated with higher self-renewal activity. Freshly prepared mouse embryonic fibroblasts (MEFs) of Rb(-/-) ; Trp53(-/-) background formed stem cell-like spheres that expressed significant levels of embryonic genes despite of lacking the ability to form colonies on soft agar or tumors in immune-deficient mice. This suggested that Rb-p53 double inactivation resulted in an undifferentiated status but without carcinogenic conversion. We next established Rb(-/-) ; N-ras(-/-) MEFs that harbored a spontaneous carcinogenic mutation in Trp53. These cells (RN6), in an Rb-dependent manner, efficiently generated spheres that expressed very high levels of embryonic genes, and appeared to be carcinogenic. We then screened an FDA-approved drug library to search for agents that suppressed the spherogenic activity of RN6 cells. Data revealed that RN6 cells were sensitive to specific agents including ones those are effective against cancer stem cells. Taken together, all these findings suggest that the genetic interaction between Rb and p53 is a critical determinant of the undifferentiated state in normal and tumor cells.


Subject(s)
Cell Differentiation , Embryo, Mammalian/cytology , Fibroblasts/cytology , Neuroendocrine Cells/cytology , Retinoblastoma Protein/metabolism , Thyroid Gland/cytology , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Behavior, Animal , Cell Line , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Heterozygote , Mice, Knockout , Molecular Sequence Data , Mutation/genetics , Phenotype , Retinoblastoma Protein/deficiency , Spheroids, Cellular/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...