Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 456
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 54(2): 367-386.e8, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33567262

ABSTRACT

Understanding the contribution of the host's genetic background to cancer immunity may lead to improved stratification for immunotherapy and to the identification of novel therapeutic targets. We investigated the effect of common and rare germline variants on 139 well-defined immune traits in ∼9000 cancer patients enrolled in TCGA. High heritability was observed for estimates of NK cell and T cell subset infiltration and for interferon signaling. Common variants of IFIH1, TMEM173 (STING1), and TMEM108 were associated with differential interferon signaling and variants mapping to RBL1 correlated with T cell subset abundance. Pathogenic or likely pathogenic variants in BRCA1 and in genes involved in telomere stabilization and Wnt-ß-catenin also acted as immune modulators. Our findings provide evidence for the impact of germline genetics on the composition and functional orientation of the tumor immune microenvironment. The curated datasets, variants, and genes identified provide a resource toward further understanding of tumor-immune interactions.


Subject(s)
Germ-Line Mutation/genetics , Immunotherapy/methods , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Genes, BRCA1 , Genome-Wide Association Study , Humans , Interferons/metabolism , Male , Middle Aged , Neoplasms/genetics , Quantitative Trait, Heritable , Retinoblastoma-Like Protein p107/genetics , Signal Transduction/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
2.
Mol Cell ; 74(4): 758-770.e4, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30982746

ABSTRACT

The cyclin-dependent kinases Cdk4 and Cdk6 form complexes with D-type cyclins to drive cell proliferation. A well-known target of cyclin D-Cdk4,6 is the retinoblastoma protein Rb, which inhibits cell-cycle progression until its inactivation by phosphorylation. However, the role of Rb phosphorylation by cyclin D-Cdk4,6 in cell-cycle progression is unclear because Rb can be phosphorylated by other cyclin-Cdks, and cyclin D-Cdk4,6 has other targets involved in cell division. Here, we show that cyclin D-Cdk4,6 docks one side of an alpha-helix in the Rb C terminus, which is not recognized by cyclins E, A, and B. This helix-based docking mechanism is shared by the p107 and p130 Rb-family members across metazoans. Mutation of the Rb C-terminal helix prevents its phosphorylation, promotes G1 arrest, and enhances Rb's tumor suppressive function. Our work conclusively demonstrates that the cyclin D-Rb interaction drives cell division and expands the diversity of known cyclin-based protein docking mechanisms.


Subject(s)
Cell Proliferation/genetics , Cyclin D/genetics , Protein Interaction Maps/genetics , Retinoblastoma Protein/genetics , Cell Cycle/genetics , Crk-Associated Substrate Protein/genetics , Cyclin D/chemistry , Cyclin-Dependent Kinase 4/chemistry , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/chemistry , Cyclin-Dependent Kinase 6/genetics , Cyclins/genetics , G1 Phase/genetics , Humans , Molecular Docking Simulation , Phosphorylation/genetics , Protein Binding/genetics , Protein Conformation, alpha-Helical/genetics , Retinoblastoma Protein/chemistry , Retinoblastoma-Like Protein p107/genetics , S Phase/genetics
3.
Proc Natl Acad Sci U S A ; 121(28): e2322917121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38959035

ABSTRACT

Functional analysis in mouse models is necessary to establish the involvement of a set of genetic variations in tumor development. A modeling platform to facilitate and cost-effectively analyze the role of multiple genes in carcinogenesis would be valuable. Here, we present an innovative strategy for lung mutagenesis using CRISPR/Cas9 ribonucleoproteins delivered via cationic polymers. This approach allows the simultaneous inactivation of multiple genes. We validate the effectiveness of this system by targeting a group of tumor suppressor genes, specifically Rb1, Rbl1, Pten, and Trp53, which were chosen for their potential to cause lung tumors, namely small cell lung carcinoma (SCLC). Tumors with histologic and transcriptomic features of human SCLC emerged after intratracheal administration of CRISPR/polymer nanoparticles. These tumors carried loss-of-function mutations in all four tumor suppressor genes at the targeted positions. These findings were reproduced in two different pure genetic backgrounds. We provide a proof of principle for simplified modeling of lung tumorigenesis to facilitate functional testing of potential cancer-related genes.


Subject(s)
CRISPR-Cas Systems , Lung Neoplasms , Mutagenesis , PTEN Phosphohydrolase , Small Cell Lung Carcinoma , Tumor Suppressor Protein p53 , Animals , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , PTEN Phosphohydrolase/genetics , Tumor Suppressor Protein p53/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Humans , Disease Models, Animal , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Gene Editing/methods
4.
Proc Natl Acad Sci U S A ; 117(1): 513-521, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871154

ABSTRACT

Small cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer that remains among the most lethal of solid tumor malignancies. Recent genomic sequencing studies have identified many recurrently mutated genes in human SCLC tumors. However, the functional roles of most of these genes remain to be validated. Here, we have adapted the CRISPR-Cas9 system to a well-established murine model of SCLC to rapidly model loss-of-function mutations in candidate genes identified from SCLC sequencing studies. We show that loss of the gene p107 significantly accelerates tumor progression. Notably, compared with loss of the closely related gene p130, loss of p107 results in fewer but larger tumors as well as earlier metastatic spread. In addition, we observe differences in proliferation and apoptosis as well as altered distribution of initiated tumors in the lung, resulting from loss of p107 or p130 Collectively, these data demonstrate the feasibility of using the CRISPR-Cas9 system to model loss of candidate tumor suppressor genes in SCLC, and we anticipate that this approach will facilitate efforts to investigate mechanisms driving tumor progression in this deadly disease.


Subject(s)
Gene Editing/methods , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Animals , Apoptosis/genetics , CRISPR-Cas Systems/genetics , Cell Line , Cell Proliferation/genetics , Disease Models, Animal , Disease Progression , Feasibility Studies , Humans , Loss of Function Mutation , Lung/pathology , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Staging , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p130/genetics , Small Cell Lung Carcinoma/pathology , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics
5.
Genes Dev ; 29(9): 961-74, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25917549

ABSTRACT

The DREAM complex represses cell cycle genes during quiescence through scaffolding MuvB proteins with E2F4/5 and the Rb tumor suppressor paralog p107 or p130. Upon cell cycle entry, MuvB dissociates from p107/p130 and recruits B-Myb and FoxM1 for up-regulating mitotic gene expression. To understand the biochemical mechanisms underpinning DREAM function and regulation, we investigated the structural basis for DREAM assembly. We identified a sequence in the MuvB component LIN52 that binds directly to the pocket domains of p107 and p130 when phosphorylated on the DYRK1A kinase site S28. A crystal structure of the LIN52-p107 complex reveals that LIN52 uses a suboptimal LxSxExL sequence together with the phosphate at nearby S28 to bind the LxCxE cleft of the pocket domain with high affinity. The structure explains the specificity for p107/p130 over Rb in the DREAM complex and how the complex is disrupted by viral oncoproteins. Based on insights from the structure, we addressed how DREAM is disassembled upon cell cycle entry. We found that p130 and B-Myb can both bind the core MuvB complex simultaneously but that cyclin-dependent kinase phosphorylation of p130 weakens its association. Together, our data inform a novel target interface for studying MuvB and p130 function and the design of inhibitors that prevent tumor escape in quiescence.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Cell Cycle Proteins/metabolism , Crystallization , Humans , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Retinoblastoma-Like Protein p107/chemistry , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/metabolism , Sequence Alignment , Trans-Activators/metabolism
6.
Proc Natl Acad Sci U S A ; 116(44): 22300-22306, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611390

ABSTRACT

High-grade neuroendocrine lung malignancies (large-cell neuroendocrine cell carcinoma, LCNEC, and small-cell lung carcinoma, SCLC) are among the most deadly lung cancer conditions with no optimal clinical management. The biological relationships between SCLC and LCNEC are still largely unknown and a current matter of debate as growing molecular data reveal high heterogeneity with potential therapeutic consequences. Here we describe murine models of high-grade neuroendocrine lung carcinomas generated by the loss of 4 tumor suppressors. In an Rbl1-null background, deletion of Rb1, Pten, and Trp53 floxed alleles after Ad-CMVcre infection in a wide variety of lung epithelial cells produces LCNEC. Meanwhile, inactivation of these genes using Ad-K5cre in basal cells leads to the development of SCLC, thus differentially influencing the lung cancer type developed. So far, a defined model of LCNEC has not been reported. Molecular and transcriptomic analyses of both models revealed strong similarities to their human counterparts. In addition, a 68Ga-DOTATOC-based molecular-imaging method provides a tool for detection and monitoring the progression of the cancer. These data offer insight into the biology of SCLC and LCNEC, providing a useful framework for development of compounds and preclinical investigations in accurate immunocompetent models.


Subject(s)
Carcinoma, Small Cell/genetics , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Neuroendocrine Tumors/genetics , Animals , Carcinoma, Small Cell/diagnostic imaging , Carcinoma, Small Cell/pathology , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Mice , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/pathology , Octreotide/analogs & derivatives , Organometallic Compounds , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Positron-Emission Tomography , Radiopharmaceuticals , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Nucleic Acids Res ; 47(21): 11197-11208, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31667499

ABSTRACT

Cell cycle gene expression occurs in two waves. The G1/S genes encode factors required for DNA synthesis and the G2/M genes contribute to mitosis. The Retinoblastoma protein (RB) and DREAM complex (DP, RB-like, E2F4 and MuvB) cooperate to repress all cell cycle genes during G1 and inhibit entry into the cell cycle. DNA damage activates p53 leading to increased levels of p21 and inhibition of cell cycle progression. Whether the G1/S and G2/M genes are differentially repressed by RB and the RB-like proteins p130 and p107 in response to DNA damage is not known. We performed gene expression profiling of primary human fibroblasts upon DNA damage and assessed the effects on G1/S and G2/M genes. Upon p53 activation, p130 and RB cooperated to repress the G1/S genes. In addition, in the absence of RB and p130, p107 contributed to repression of G1/S genes. In contrast, G2/M genes were repressed by p130 and p107 after p53 activation. Furthermore, repression of G2/M genes by p107 and p130 led to reduced entry into mitosis. Our data demonstrates specific roles for RB, p130-DREAM, and p107-DREAM in p53 and p21 mediated repression of cell cycle genes.


Subject(s)
G1 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p130/genetics , Tumor Suppressor Protein p53/metabolism , Cell Cycle/genetics , Cells, Cultured , Down-Regulation/genetics , Genes, cdc/genetics , Humans , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/metabolism , Tumor Suppressor Protein p53/genetics
8.
Nucleic Acids Res ; 47(17): 9087-9103, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31400114

ABSTRACT

Most human cancers acquire mutations causing defects in the p53 signaling pathway. The tumor suppressor p53 becomes activated in response to genotoxic stress and is essential for arresting the cell cycle to facilitate DNA repair or to initiate apoptosis. p53-induced cell cycle-arrest is mediated by expression of the CDK inhibitor p21WAF1/Cip1, which prevents phosphorylation and inactivation of the pocket proteins RB, p130, and p107. In a hypophosphorylated state, pocket proteins bind to E2F factors forming RB-E2F and DREAM transcriptional repressor complexes. Here, we analyze the influence of RB and DREAM on p53-induced gene repression and cell-cycle arrest. We show that abrogation of DREAM function by knockout of the DREAM component LIN37 results in a reduced repression of cell-cycle genes. We identify the genes repressed by the p53-DREAM pathway and describe a set of genes that is downregulated by p53 independent of LIN37/DREAM. Most strikingly, p53-dependent repression of cell-cycle genes is completely abrogated in LIN37-/-;RB-/- cells leading to a loss of the G1/S checkpoint. Taken together, we show that DREAM and RB are key factors in the p53 signaling pathway to downregulate a large number of cell-cycle genes and to arrest the cell cycle at the G1/S transition.


Subject(s)
Cell Cycle Checkpoints/genetics , Gene Expression Regulation , Kv Channel-Interacting Proteins/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/genetics , Trans-Activators/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Crk-Associated Substrate Protein/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Fibroblasts/metabolism , Genes, cdc , HCT116 Cells , Humans , Kv Channel-Interacting Proteins/genetics , Mice , Repressor Proteins/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Suppressor Protein p53/genetics
9.
Nature ; 514(7522): 385-8, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25252974

ABSTRACT

Retinoblastoma is a childhood retinal tumour that initiates in response to biallelic RB1 inactivation and loss of functional retinoblastoma (Rb) protein. Although Rb has diverse tumour-suppressor functions and is inactivated in many cancers, germline RB1 mutations predispose to retinoblastoma far more strongly than to other malignancies. This tropism suggests that retinal cell-type-specific circuitry sensitizes to Rb loss, yet the nature of the circuitry and the cell type in which it operates have been unclear. Here we show that post-mitotic human cone precursors are uniquely sensitive to Rb depletion. Rb knockdown induced cone precursor proliferation in prospectively isolated populations and in intact retina. Proliferation followed the induction of E2F-regulated genes, and depended on factors having strong expression in maturing cone precursors and crucial roles in retinoblastoma cell proliferation, including MYCN and MDM2. Proliferation of Rb-depleted cones and retinoblastoma cells also depended on the Rb-related protein p107, SKP2, and a p27 downregulation associated with cone precursor maturation. Moreover, Rb-depleted cone precursors formed tumours in orthotopic xenografts with histological features and protein expression typical of human retinoblastoma. These findings provide a compelling molecular rationale for a cone precursor origin of retinoblastoma. More generally, they demonstrate that cell-type-specific circuitry can collaborate with an initiating oncogenic mutation to enable tumorigenesis.


Subject(s)
Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinoblastoma Protein/metabolism , Retinoblastoma/metabolism , Retinoblastoma/pathology , Cell Transformation, Neoplastic , E2F Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Genes, Retinoblastoma/genetics , Heterografts , Humans , N-Myc Proto-Oncogene Protein , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Organ Specificity , Proto-Oncogene Proteins c-mdm2/metabolism , Retinoblastoma/genetics , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/deficiency , Retinoblastoma-Like Protein p130/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Stem Cells/metabolism , Stem Cells/pathology
10.
Proc Natl Acad Sci U S A ; 114(19): 4942-4947, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28439018

ABSTRACT

The retinoblastoma protein (Rb) and the homologous pocket proteins p107 and p130 negatively regulate cell proliferation by binding and inhibiting members of the E2F transcription factor family. The structural features that distinguish Rb from other pocket proteins have been unclear but are critical for understanding their functional diversity and determining why Rb has unique tumor suppressor activities. We describe here important differences in how the Rb and p107 C-terminal domains (CTDs) associate with the coiled-coil and marked-box domains (CMs) of E2Fs. We find that although CTD-CM binding is conserved across protein families, Rb and p107 CTDs show clear preferences for different E2Fs. A crystal structure of the p107 CTD bound to E2F5 and its dimer partner DP1 reveals the molecular basis for pocket protein-E2F binding specificity and how cyclin-dependent kinases differentially regulate pocket proteins through CTD phosphorylation. Our structural and biochemical data together with phylogenetic analyses of Rb and E2F proteins support the conclusion that Rb evolved specific structural motifs that confer its unique capacity to bind with high affinity those E2Fs that are the most potent activators of the cell cycle.


Subject(s)
E2F Transcription Factors/chemistry , Retinoblastoma Protein/chemistry , Retinoblastoma-Like Protein p107/chemistry , Crystallography, X-Ray , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Humans , Protein Domains , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism
11.
J Mammary Gland Biol Neoplasia ; 24(2): 163-175, 2019 06.
Article in English | MEDLINE | ID: mdl-30613869

ABSTRACT

High mammographic density (MD) is associated with a 4-6 times increase in breast cancer risk. For post-menopausal women, MD often decreases over time, but little is known about the underlying biological mechanisms. MD reflects breast tissue composition, and may be associated with microenvironment subtypes previously identified in tumor-adjacent normal tissue. Currently, these subtypes have not been explored in normal breast tissue. We obtained biopsies from breasts of healthy women at two different time points several years apart and performed microarray gene expression analysis. At time point 1, 65 samples with both MD and gene expression were available. At time point 2, gene expression and MD data were available from 17 women, of which 11 also had gene expression data available from the first time point. We validated findings from our previous study; negative correlation between RBL1 and MD in post-menopausal women, indicating involvement of the TGFß pathway. We also found that breast tissue samples from women with a large decrease in MD sustained higher expression of genes in the histone family H4. In addition, we explored the previously defined active and inactive microenvironment subtypes and demonstrated that normal breast samples of the active subtype had characteristics similar to the claudin-low breast cancer subtype. Breast biopsies from healthy women are challenging to obtain, but despite a limited sample size, we have identified possible mechanisms relevant for changes in breast biology and MD over time that may be of importance for breast cancer risk and tumor initiation.


Subject(s)
Breast Density/genetics , Breast Neoplasms/diagnosis , Breast/diagnostic imaging , Retinoblastoma-Like Protein p107/genetics , Aged , Biomarkers/metabolism , Biopsy , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Datasets as Topic , Female , Gene Expression Profiling , Histones/genetics , Histones/metabolism , Humans , Longitudinal Studies , Mammography , Middle Aged , Retinoblastoma-Like Protein p107/metabolism , Tumor Microenvironment/genetics
12.
Am J Physiol Endocrinol Metab ; 316(1): E135-E144, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30512986

ABSTRACT

During pregnancy, maternal pancreatic ß-cells undergo a compensatory expansion in response to the state of insulin resistance, where prolactin (PRL) plays a major role. Retinoblastoma protein (Rb) has been shown to critically regulate islet proliferation and function. The aim of the study was to explore the role of Rb in ß-cell mass expansion during pregnancy. Expression of pocket protein family and E2Fs were examined in mouse islets during pregnancy and in insulinoma cells (INS-1) stimulated by PRL. PRL-stimulated INS-1 cells were used to explore the signaling pathway that regulates Rb downstream of the PRL receptor. Pancreas-specific Rb-knockout (Rb-KO) mice were assessed to evaluate the in vivo function of Rb in ß-cell proliferation during pregnancy. During pregnancy, expression of Rb, phospho-Rb (p-Rb), p107, and E2F1 increased, while p130 decreased in maternal islets. With PRL stimulation, induction of Rb expression occurred mainly in the nucleus, while p-Rb was predominantly in the cytoplasm. Inhibition of STAT5 significantly restrained the expression of CDK4, Rb, p-Rb, and E2F1 in PRL-stimulated INS-1 cells with attenuation in cell cycle progression. Reduction of Rb phosphorylation by CDK4 inhibition blocked PRL-mediated proliferation of INS-1 cells. On the other hand, knockdown of Rb using siRNA led to an induction in E2F1 leading to cell cycle progression from G1 to S and G2/M phase, similar to the effects of PRL-mediated induction of p-Rb that led to cell proliferation. With Rb knockdown, PRL did not lead to further increase in cell cycle progression. Similarly, while Rb-KO pregnant mice displayed better glucose tolerance and higher insulin secretion, they had similar ß-cell mass and proliferation to wild-type pregnant controls, supporting the essential role of Rb suppression in augmenting ß-cell proliferation during pregnancy. Rb-E2F1 regulation plays a pivotal role in PRL-stimulated ß-cell proliferation. PRL promotes Rb phosphorylation and E2F1 upregulation via STAT5-cyclin D/CDK4 pathway during pregnancy.


Subject(s)
Cell Proliferation/genetics , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Insulin-Secreting Cells/metabolism , Pregnancy/metabolism , Retinoblastoma Protein/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , E2F1 Transcription Factor/metabolism , Female , Insulin-Secreting Cells/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Prolactin/pharmacology , Rats , Retinoblastoma-Like Protein p107/metabolism , STAT5 Transcription Factor
13.
Genes Dev ; 25(16): 1734-45, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21816922

ABSTRACT

The miR-17~92 cluster is a potent microRNA-encoding oncogene. Here, we show that miR-17~92 synergizes with loss of Rb family members to promote retinoblastoma. We observed miR-17~92 genomic amplifications in murine retinoblastoma and high expression of miR-17~92 in human retinoblastoma. While miR-17~92 was dispensable for mouse retinal development, miR-17~92 overexpression, together with deletion of Rb and p107, led to rapid emergence of retinoblastoma with frequent metastasis to the brain. miR-17~92 oncogenic function in retinoblastoma was not mediated by a miR-19/PTEN axis toward apoptosis suppression, as found in lymphoma/leukemia models. Instead, miR-17~92 increased the proliferative capacity of Rb/p107-deficient retinal cells. We found that deletion of Rb family members led to compensatory up-regulation of the cyclin-dependent kinase inhibitor p21Cip1. miR-17~92 overexpression counteracted p21Cip1 up-regulation, promoted proliferation, and drove retinoblastoma formation. These results demonstrate that the oncogenic determinants of miR-17~92 are context-specific and provide new insights into miR-17~92 function as an RB-collaborating gene in cancer.


Subject(s)
MicroRNAs/genetics , Mutation , Retinoblastoma Protein/genetics , Retinoblastoma/genetics , Signal Transduction/genetics , Animals , Animals, Newborn , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Multigene Family , Oligonucleotide Array Sequence Analysis , Oligonucleotides, Antisense/genetics , Pregnancy , Retina/embryology , Retina/growth & development , Retina/metabolism , Retinoblastoma/metabolism , Retinoblastoma/pathology , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism
14.
J Biol Chem ; 292(16): 6583-6599, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28289097

ABSTRACT

The human cytomegalovirus (HCMV)-encoded viral cyclin-dependent kinase (v-CDK) UL97 phosphorylates the retinoblastoma (Rb) tumor suppressor. Here, we identify the other Rb family members p107 and p130 as novel targets of UL97. UL97 phosphorylates p107 and p130 thereby inhibiting their ability to repress the E2F-responsive E2F1 promoter. As with Rb, this phosphorylation, and the rescue of E2F-responsive transcription, is dependent on the L1 LXCXE motif in UL97 and its interacting clefts on p107 and p130. Interestingly, UL97 does not induce the disruption of all p107-E2F or p130-E2F complexes, as it does to Rb-E2F complexes. UL97 strongly interacts with p107 but not Rb or p130. Thus the inhibitory mechanisms of UL97 for Rb family protein-mediated repression of E2F-responsive transcription appear to differ for each of the Rb family proteins. The immediate early 1 (IE1) protein of HCMV also rescues p107- and p130-mediated repression of E2F-responsive gene expression, but it does not induce their phosphorylation and does not disrupt p107-E2F or p130-E2F complexes. The unique regulation of Rb family proteins by HCMV UL97 and IE1 attests to the importance of modulating Rb family protein function in HCMV-infected cells.


Subject(s)
Cytomegalovirus Infections/metabolism , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/metabolism , Alleles , Amino Acid Motifs , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Immediate-Early Proteins/metabolism , Mutation , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Promoter Regions, Genetic
15.
Stem Cells ; 35(5): 1378-1391, 2017 05.
Article in English | MEDLINE | ID: mdl-28233396

ABSTRACT

We show that the transcriptional corepressor p107 orchestrates a metabolic checkpoint that determines adipocyte lineage fates for non-committed progenitors. p107 accomplishes this when stem cell commitment would normally occur in growth arrested cells. p107-deficient embryonic progenitors are characterized by a metabolic state resembling aerobic glycolysis that is necessary for their pro-thermogenic fate. Indeed, during growth arrest they have a reduced capacity for NADH partitioning between the cytoplasm and mitochondria. Intriguingly, this occurred despite an increase in the capacity for mitochondrial oxidation of non-glucose substrates. The significance of metabolic reprogramming is underscored by the disruption of glycolytic capacities in p107-depleted progenitors that reverted their fates from pro-thermogenic to white adipocytes. Moreover, the manipulation of glycolytic capacity on nonspecified embryonic and adult progenitors forced their beige fat commitment. These innovative findings introduce a new approach to increase pro-thermogenic adipocytes based on simply promoting aerobic glycolysis to manipulate nonspecified progenitor fate decisions. Stem Cells 2017;35:1378-1391.


Subject(s)
Adipocytes, Brown/cytology , Adipocytes, White/cytology , Cell Cycle Checkpoints , Cell Lineage , Retinoblastoma-Like Protein p107/metabolism , Aerobiosis , Animals , Cell Line , Cell Nucleus/metabolism , Embryo, Mammalian/cytology , Gene Knockdown Techniques , Glycolysis , Mice, Knockout , Models, Biological , Oxidation-Reduction , Stem Cells/cytology , Stem Cells/metabolism , Subcellular Fractions/metabolism
16.
Genes Dev ; 24(13): 1377-88, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20551164

ABSTRACT

Loss of G1/S control is a hallmark of cancer, and is often caused by inactivation of the retinoblastoma pathway. However, mouse embryonic fibroblasts lacking the retinoblastoma genes RB1, p107, and p130 (TKO MEFs) are still subject to cell cycle control: Upon mitogen deprivation, they enter and complete S phase, but then firmly arrest in G2. We now show that G2-arrested TKO MEFs have accumulated DNA damage. Upon mitogen readdition, cells resume proliferation, although only part of the damage is repaired. As a result, mitotic cells show chromatid breaks and chromatid cohesion defects. These aberrations lead to aneuploidy in the descendent cell population. Thus, our results demonstrate that unfavorable growth conditions can cause genomic instability in cells lacking G1/S control. This mechanism may allow premalignant tumor cells to acquire additional genetic alterations that promote tumorigenesis.


Subject(s)
Genomic Instability , Mitogens/physiology , Retinoblastoma Protein , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Signal Transduction/physiology , Aneuploidy , Animals , Cell Cycle , Cell Proliferation/drug effects , Centromere , DNA Breaks, Double-Stranded , DNA Copy Number Variations , Fibroblasts/cytology , Mice , Mitogens/pharmacology , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/deficiency , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/deficiency , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism
17.
Genes Chromosomes Cancer ; 56(3): 231-242, 2017 03.
Article in English | MEDLINE | ID: mdl-27750399

ABSTRACT

Several murine retinoblastoma models have been generated by deleting the genes encoding for retinoblastoma susceptibility protein pRb and one of its family members p107 or p130. In Rb-/- p107-/- retinoblastomas, somatic copy number alterations (SCNAs) like Mdm2 amplification or Cdkn2a deletion targeting the p53-pathway occur, which is uncommon for human retinoblastoma. In our study, we determined SCNAs in retinoblastomas developing in Rb-/- p130-/- mice and compared this to murine Rb-/- p107-/- tumors and human tumors. Chimeric mice were made by injection of 129/Ola-derived Rb-/- p130-/- embryonic stem cells into wild type C57BL/6 blastocysts. SCNAs of retinoblastoma samples were determined by low-coverage (∼0.5×) whole genome sequencing. In Rb-/- p130-/- tumors, SCNAs included gain of chromosomes 1 (3/23 tumors), 8 (1/23 tumors), 10 (1/23 tumors), 11 (2/23 tumors), and 12 (4/23 tumors), which could be mapped to frequently altered chromosomes in human retinoblastomas. While the altered chromosomes in Rb-/- p130-/- tumors were similar to those in Rb-/- p107-/- tumors, the alteration frequencies were much lower in Rb-/- p130-/- tumors. Most of the Rb-/- p130-/- tumors (16/23 tumors, 70%) were devoid of SCNAs, in strong contrast to Rb-/- p107-/- tumors, which were never (0/15 tumors) SCNA-devoid. Similarly, to human retinoblastoma, increased age at diagnosis significantly correlated with increased SCNA frequencies. Additionally, focal loss of Cdh11 was observed in one Rb-/- p130-/- tumor, which enforces studies in human retinoblastoma that identified CDH11 as a retinoblastoma suppressor. Moreover, based on a comparison of genes altered in human and murine retinoblastoma, we suggest exploring the role of HMGA1 and SRSF3 in retinoblastoma development. © 2016 Wiley Periodicals, Inc.


Subject(s)
Biomarkers, Tumor/genetics , DNA Copy Number Variations/genetics , Retinoblastoma-Like Protein p107/physiology , Retinoblastoma-Like Protein p130/physiology , Retinoblastoma/genetics , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Genome , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Mice , Mice, Inbred C57BL
18.
Microvasc Res ; 114: 26-33, 2017 11.
Article in English | MEDLINE | ID: mdl-28595801

ABSTRACT

miRNAs are important regulators of cellular senescence yet the extent of their involvement remains to be investigated. We sought to identify miRNAs that are involved in cytokine-induced premature senescence (CIPS) in endothelial cells. CIPS was established in young human pulmonary microvascular endothelial cells (HMVEC-Ls) following treatment with a sublethal dose (20ng/ml) of tumor necrosis factor alpha (TNF-α) for 15days. In parallel, HMVEC-Ls were grown and routinely passaged until the onset of replicative senescence (RS). Differential expression analysis following miRNA microarray profiling revealed an overlapped of eight deregulated miRNAs in both the miRNA profiles of RS and TNF-α-induced premature senescence cells. Amongst the deregulated miRNAs were members of the miR 17-92 cluster which are known regulators of angiogenesis. The role of hsa-miR-20b in TNF-α-induced premature senescence, a paralog member of the miR 17-92 cluster, was further investigated. Biotin-labeled hsa-miR-20b captured the enriched transcripts of retinoblastoma-like 1 (RBL1), indicating that RBL1 is a target of hsa-miR-20b. Knockdown of hsa-miR-20b attenuated premature senescence in the TNF-α-treated HMVEC-Ls as evidenced by increased cell proliferation, increased RBL1 mRNA expression level but decreased protein expression of p16INK4a, a cellular senescence marker. These findings provide an early insight into the role of hsa-miR-20b in endothelial senescence.


Subject(s)
Cell Proliferation/drug effects , Cellular Senescence/drug effects , Endothelial Cells/drug effects , Lung/blood supply , MicroRNAs/metabolism , Microvessels/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Humans , MicroRNAs/genetics , Microvessels/metabolism , Microvessels/pathology , RNA Interference , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Time Factors , Transfection
19.
Biologicals ; 46: 143-147, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28222938

ABSTRACT

The miR-17-92 cluster consisted of seven miRNAs (mir-17-5p, -17-3p, -18a, -19a, -20a, -19b-1, and -92a-1). Previous studies have shown this cluster has been over-expressed in several cancers. The aim of this study was to evaluate the over-expression impacts of miR-17-92 on stem cells. In the current work, the effect of miR-17-92 cluster which was cloned in Lentiviral vector has been investigated on unrestricted somatic stem cells (USSCs). Tumor suppressor genes (p53, p15, RBL1, SMAD2, SMAD4, and MAPK-1) expression, especially p53, was considerably reduced. These data show the potential of miR-17-92 for oncogenesis regulation in stem cells. In conclusion, the role of miR-17-92 in USSCs may provide a better understanding of its function in tumorigenesis and for the possible use in cell therapy of the anti-mir-17-92 cluster.


Subject(s)
Gene Expression Regulation , Genes, Tumor Suppressor , Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , Multigene Family , Cell Cycle/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15/genetics , Fetal Blood/cytology , Humans , Mitogen-Activated Protein Kinase 1/genetics , RNA, Long Noncoding , Retinoblastoma-Like Protein p107/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/genetics , Smad4 Protein/genetics , Tumor Suppressor Protein p53/genetics
20.
Int J Mol Sci ; 18(8)2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28812991

ABSTRACT

The retinoblastoma (RB) tumor suppressor is known as a master regulator of the cell cycle. RB is mutated or functionally inactivated in the majority of human cancers. This transcriptional regulator exerts its function in cell cycle control through its interaction with the E2F family of transcription factors and with chromatin remodelers and modifiers that contribute to the repression of genes important for cell cycle progression. Over the years, studies have shown that RB participates in multiple processes in addition to cell cycle control. Indeed, RB is known to interact with over 200 different proteins and likely exists in multiple complexes. RB, in some cases, acts through its interaction with E2F1, other members of the pocket protein family (p107 and p130), and/or chromatin remodelers and modifiers. RB is a tumor suppressor with important chromatin regulatory functions that affect genomic stability. These functions include the role of RB in DNA repair, telomere maintenance, chromosome condensation and cohesion, and silencing of repetitive regions. In this review we will discuss recent advances in RB biology related to RB, partner proteins, and their non-transcriptional functions fighting back against genomic instability.


Subject(s)
DNA Repair , Gene Silencing , Genomic Instability , Retinoblastoma Protein/metabolism , Telomere Homeostasis , Animals , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Humans , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL