Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
J Immunother Cancer ; 10(3)2022 03.
Article En | MEDLINE | ID: mdl-35296559

BACKGROUND: Bispecific T-cell engager (BiTE) molecules induce redirected lysis of cancer cells by T cells and are an emerging modality for solid tumor immunotherapy. While signs of clinical activity have been demonstrated, efficacy of T-cell engagers (TCEs) in solid tumors settings, molecular determinants of response, and underlying mechanisms of resistance to BiTE therapy require more investigation. METHODS: To uncover cancer cell-intrinsic genetic modifiers of TCE-mediated cytotoxicity, we performed genome-wide CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) loss-of-function and CRISPRa (CRISPR activation) gain-of-function screens using TCEs against two distinct tumor-associated antigens (TAAs). By using in vitro T-cell cytotoxicity assays and in vivo efficacy studies, we validated the roles of two common pathways identified in our screen, T-cell costimulation pathway and apoptosis pathway, as key modifiers of BiTE activity. RESULTS: Our genetic screens uncovered TAAs-independent cancer cell-intrinsic genes with functions in autophagy, T-cell costimulation, the apoptosis pathway, chromatin remodeling, and cytokine signaling that altered responsiveness to BiTE-mediated killing. Notably, loss of CD58 (the ligand of the CD2 T-cell costimulatory receptor), a gene frequently altered in cancer, led to decreased TCE-mediated cytotoxicity, T-cell activation and antitumor efficacy in vitro and in vivo. Moreover, the effects of CD58 loss were synergistically compounded by concurrent loss of CD80/CD86 (ligands for the CD28 T-cell costimulatory receptor), whereas joint CD2 and CD28 costimulation additively enhanced TCE-mediated killing, indicating non-redundant costimulatory mechanisms between the two pathways. Additionally, loss of CFLAR (Caspase-8 and FADD Like Apoptosis Regulator), BCL2L1, and BID (BH3 Interacting Domain Death Agonist) induced profound changes in sensitivity to TCEs, indicating that key regulators of apoptosis, which are frequently altered in cancer, impact tumor responsiveness to BiTE therapy. CONCLUSIONS: This study demonstrates that genetic alterations central to carcinogenesis and commonly detected in cancer samples lead to significant modulation of BiTE antitumor activity in vitro and in vivo, findings with relevance for a better understanding of patient responses to BiTE therapy and novel combinations that enhance TCE efficacy.


CD58 Antigens/immunology , Immunotherapy , Neoplasms , Apoptosis , CD28 Antigens/metabolism , Humans , Lymphocyte Activation , Neoplasms/metabolism , T-Lymphocytes
2.
J Mol Biol ; 432(16): 4544-4560, 2020 07 24.
Article En | MEDLINE | ID: mdl-32534063

The regulation of gene expression is a result of a complex interplay between chromatin remodeling, transcription factors, and signaling molecules. Cell differentiation is accompanied by chromatin remodeling of specific loci to permanently silence genes that are not essential for the differentiated cell activity. The molecular cues that recruit the chromatin remodeling machinery are not well characterized. IRF8 is an immune-cell specific transcription factor and its expression is augmented by interferon-γ. Therefore, it serves as a model gene to elucidate the molecular mechanisms governing its silencing in non-immune cells. Ahigh-throughput shRNA library screen in IRF8 expression-restrictive cells enabled the identification of MafK as modulator of IRF8 silencing, affecting chromatin architecture. ChIP-Seq analysis revealed three MafK binding regions (-25 kb, -20 kb, and IRF8 6th intron) within the IRF8 locus. These MafK binding sites are sufficient to repress a reporter gene when cloned in genome-integrated lentiviral reporter constructs in only expression-restrictive cells. Conversely, plasmid-based constructs do not demonstrate such repressive effect. These results highlight the role of these MafK binding sites in mediating repressed chromatin assembly. Finally, a more thorough genomic analysis was performed, using CRISPR-Cas9 to delete MafK-int6 binding region in IRF8 expression-restrictive cells. Deleted clones exhibited an accessible chromatin conformation within the IRF8 locus that was accompanied by a significant increase in basal expression of IRF8 that was further induced by interferon-γ. Taken together, we identified and characterized several MafK binding elements within the IRF8 locus that mediate repressive chromatin conformation resulting in the silencing of IRF8 expression in a celltype-specific manner.


Chromatin/metabolism , Interferon Regulatory Factors/genetics , MafK Transcription Factor/genetics , MafK Transcription Factor/metabolism , Animals , Binding Sites , CRISPR-Cas Systems , Chromatin Assembly and Disassembly , Gene Expression Regulation , HEK293 Cells , Humans , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/metabolism , Mice , NIH 3T3 Cells , Organ Specificity , RAW 264.7 Cells , RNA, Small Interfering/pharmacology
3.
Cancer Res ; 79(12): 3125-3138, 2019 06 15.
Article En | MEDLINE | ID: mdl-31000522

Oncogenic MYC activation promotes proliferation in Burkitt lymphoma, but also induces cell-cycle arrest and apoptosis mediated by p53, a tumor suppressor that is mutated in 40% of Burkitt lymphoma cases. To identify molecular dependencies in Burkitt lymphoma, we performed RNAi-based, loss-of-function screening in eight Burkitt lymphoma cell lines and integrated non-Burkitt lymphoma RNAi screens and genetic data. We identified 76 genes essential to Burkitt lymphoma, including genes associated with hematopoietic cell differentiation (FLI1, BCL11A) or B-cell development and activation (PAX5, CDKN1B, JAK2, CARD11) and found a number of context-specific dependencies including oncogene addiction in cell lines with TCF3/ID3 or MYD88 mutation. The strongest genotype-phenotype association was seen for TP53. MDM4, a negative regulator of TP53, was essential in TP53 wild-type (TP53wt) Burkitt lymphoma cell lines. MDM4 knockdown activated p53, induced cell-cycle arrest, and decreased tumor growth in a xenograft model in a p53-dependent manner. Small molecule inhibition of the MDM4-p53 interaction was effective only in TP53wt Burkitt lymphoma cell lines. Moreover, primary TP53wt Burkitt lymphoma samples frequently acquired gains of chromosome 1q, which includes the MDM4 locus, and showed elevated MDM4 mRNA levels. 1q gain was associated with TP53wt across 789 cancer cell lines and MDM4 was essential in the TP53wt-context in 216 cell lines representing 19 cancer entities from the Achilles Project. Our findings highlight the critical role of p53 as a tumor suppressor in Burkitt lymphoma and identify MDM4 as a functional target of 1q gain in a wide range of cancers that is therapeutically targetable. SIGNIFICANCE: Targeting MDM4 to alleviate degradation of p53 can be exploited therapeutically across Burkitt lymphoma and other cancers with wild-type p53 harboring 1q gain, the most frequent copy number alteration in cancer.


Burkitt Lymphoma/pathology , Cell Cycle Proteins/metabolism , Chromosome Aberrations , Chromosomes, Human, Pair 1/genetics , Gene Expression Regulation, Neoplastic , Mutation , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Proliferation , Humans , Mice , Proto-Oncogene Proteins/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
Drug Discov Today Technol ; 11: 11-8, 2014 Mar.
Article En | MEDLINE | ID: mdl-24847648

RNAi loss-of-function screens, which have proven effective to identify genes functionally responsible for cellular phenotypes, can be designed to use different genetic backgrounds or altered environmental conditions to elucidate genetic dependencies. These sorts of screening approaches can be exploited to identify genetic targets that minimize resistance to approved drugs, and provide a basis on which to develop new targeted therapies and predict the secondary targets for combinatorial treatments. Four types of pooled short hairpin RNA (shRNA) screens, in particular, have been used to look for genetic targets that work together with known drugs or other anticancer targets, either in an additive or synergistic fashion. Each method produces results that provide a useful but limited picture of the genetic elements driving oncogenesis.


Drug Resistance, Neoplasm , Neoplasms/genetics , RNA Interference , Gene Knockdown Techniques , Humans
5.
PLoS One ; 8(6): e67316, 2013.
Article En | MEDLINE | ID: mdl-23840661

Advances in the fields of cancer initiating cells and high-throughput in vivo shRNA screens have highlighted a need to observe the growth of tumor cells in cancer models at the clonal level. While in vivo cancer cell growth heterogeneity in xenografts has been described, it has yet to be measured. Here, we tested an approach to quantify the clonal growth heterogeneity of cancer cells in subcutaneous xenograft mouse models. Using a high-throughput sequencing method, we followed the fate in vitro and in vivo of ten thousand HCT-116 cells individually tagged with a unique barcode delivered by lentiviral transduction. While growth in vitro was less homogeneous than anticipated, we still find that 95% of the final cells derived from 80% of the original cells. In xenografts, however, 95% of the retrieved barcoded cells originated from only 6% of the initially injected cells, an effect we term "clonal dominance". We observed this clonal dominance in two additional xenograft models (MDA-MB-468 and A2780(cis)) and in two different host strains (NSG and Nude). By precisely and reproducibly quantifying clonal cancer cell growth in vivo, we find that a small subset of clones accounts for the vast majority of the descendant cells, even with HCT-116, a cell line reported to lack a tumor-initiating compartment. The stochastic in vivo selection process we describe has important implications for the fields of in vivo shRNA screening and tumor initiating cells.


Cell Transformation, Neoplastic , Lentivirus/genetics , Animals , Cell Proliferation , Clone Cells/pathology , Female , HCT116 Cells , Humans , Mice , RNA, Small Interfering/genetics , Transduction, Genetic
6.
J Cell Biol ; 178(3): 371-85, 2007 Jul 30.
Article En | MEDLINE | ID: mdl-17664332

Overexpression of cyclin E, an activator of cyclin-dependent kinase 2, has been linked to human cancer. In cell culture models, the forced expression of cyclin E leads to aneuploidy and polyploidy, which is consistent with a direct role of cyclin E overexpression in tumorigenesis. In this study, we show that the overexpression of cyclin E has a direct effect on progression through the latter stages of mitotic prometaphase before the complete alignment of chromosomes at the metaphase plate. In some cases, such cells fail to divide chromosomes, resulting in polyploidy. In others, cells proceed to anaphase without the complete alignment of chromosomes. These phenotypes can be explained by an ability of overexpressed cyclin E to inhibit residual anaphase-promoting complex (APC(Cdh1)) activity that persists as cells progress up to and through the early stages of mitosis, resulting in the abnormal accumulation of APC(Cdh1) substrates as cells enter mitosis. We further show that the accumulation of securin and cyclin B1 can account for the cyclin E-mediated mitotic phenotype.


Cell Cycle/physiology , Cyclin E/metabolism , Mitosis/physiology , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Line , Cyclin A/metabolism , Cyclin B/genetics , Cyclin B/metabolism , Cyclin B1 , Cyclin E/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Humans , Microscopy, Fluorescence/methods , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Securin , Ubiquitin/metabolism , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Neoplasia ; 9(7): 601-13, 2007 Jul.
Article En | MEDLINE | ID: mdl-17710163

We identified the ubiquitin-conjugating enzyme E2-EPF mRNA as differentially expressed in breast tumors relative to normal tissues and performed studies to elucidate its putative role in cancer. We demonstrated that overexpression of E2-EPF protein correlated with estrogen receptor (ER) negativity in breast cancer specimens and that its expression is cell cycle-regulated, suggesting a potential function for E2-EPF in cell cycle progression. However, reduction of E2-EPF protein levels by > 80% using RNAi had no significant effects on the proliferation of HeLa cervical cancer cells or ER(-) MDA-MB-231 or MDA-MB-453 breast cancer cells. Because E2-EPF protein levels were elevated during the G(2)/M phase of the cell cycle and because E2-EPF mRNA in tumor specimens was frequently coexpressed with genes involved in cell cycle control, spindle assembly, and mitotic surveillance, the possibility that E2-EPF might have a function in the cellular response to agents that induce a G(2) checkpoint or an M checkpoint was investigated. E2-EPF knockdown sensitized HeLa cells to the topoisomerase (topo) II inhibitors etoposide and doxorubicin and also increased topo IIalpha protein levels. These data suggest that combined administration of topo II-directed drugs and E2-EPF inhibitors may enhance their clinical effectiveness.


Breast Neoplasms/enzymology , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Topoisomerase II Inhibitors , Ubiquitin-Conjugating Enzymes/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Etoposide/pharmacology , HeLa Cells , Humans , RNA, Small Interfering/pharmacology , Receptors, Estrogen/metabolism , Transcriptional Activation , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/genetics , Up-Regulation
8.
Cancer Res ; 65(18): 8298-307, 2005 Sep 15.
Article En | MEDLINE | ID: mdl-16166306

In primary cells, oncogenic ras induces a stable growth arrest known as premature senescence. Ras-induced premature senescence is considered as a tumor-suppressing defense response that needs to be bypassed before oncogenic potential ras can be revealed. To gain insights into the mechanism of senescence bypass during oncogenic transformation, we dissected the activities of an adenoviral oncoprotein E1A, which is capable of overcoming ras-induced senescence. Our results have indicated that the senescence bypassing activity resides in the NH2 terminus and requires both Rb-binding and p300/CBP-binding functions of E1A. Although interference with the p16(INK4A)/Rb pathway or inactivation of p300/CBP alone did not lead to senescence bypass, these two types of genetic alterations complemented the Rb-binding defective and the p300/CBP-binding defective mutants of E1A, respectively, to rescue premature senescence. Therefore, genetic alterations disrupting the p16(INK4A)/Rb pathway or the p300/CBP functions both contribute to the bypass of senescence. We further showed that p300/CBP were essential for ras-induced p53 activity, providing a potential mechanism underlying the important role of p300/CBP in senescence. Furthermore, p300/CBP inactivation led to cellular transformation in cooperation with the p300/CBP-binding defective E1A mutants, MDM2 and Ha-RasV12. These results have shown that p300 and CBP are integral components of the pathway that mediates ras-induced senescence. The critical role of p300 and CBP in the senescence response that limits the oncogenic potential of ras has provided a mechanistic basis for the tumor-suppressing function of these proteins.


Adenovirus E1A Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Genes, ras/physiology , Retinoblastoma Protein/metabolism , p300-CBP Transcription Factors/metabolism , Adenovirus E1A Proteins/genetics , Cells, Cultured , Cellular Senescence/genetics , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mutation , Protein Binding , RNA, Small Interfering/genetics , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
9.
J Cell Biol ; 165(6): 789-800, 2004 Jun 21.
Article En | MEDLINE | ID: mdl-15197178

Deregulation of cyclin E expression has been associated with a broad spectrum of human malignancies. Analysis of DNA replication in cells constitutively expressing cyclin E at levels similar to those observed in a subset of tumor-derived cell lines indicates that initiation of replication and possibly fork movement are severely impaired. Such cells show a specific defect in loading of initiator proteins Mcm4, Mcm7, and to a lesser degree, Mcm2 onto chromatin during telophase and early G1 when Mcm2-7 are normally recruited to license origins of replication. Because minichromosome maintenance complex proteins are thought to function as a heterohexamer, loading of Mcm2-, Mcm4-, and Mcm7-depleted complexes is likely to underlie the S phase defects observed in cyclin E-deregulated cells, consistent with a role for minichromosome maintenance complex proteins in initiation of replication and fork movement. Cyclin E-mediated impairment of DNA replication provides a potential mechanism for chromosome instability observed as a consequence of cyclin E deregulation.


Cyclin E/physiology , DNA Replication/physiology , Breast Neoplasms , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin E/genetics , DNA Replication/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Flow Cytometry , G1 Phase , Humans , KB Cells , Minichromosome Maintenance Complex Component 2 , Minichromosome Maintenance Complex Component 4 , Minichromosome Maintenance Complex Component 7 , Nuclear Proteins/deficiency , Nuclear Proteins/genetics
10.
Genes Dev ; 16(22): 2946-57, 2002 Nov 15.
Article En | MEDLINE | ID: mdl-12435635

p130 is a tumor suppressor of the pocket protein family whose expression is posttranscriptionally regulated and largely G0 restricted. The mechanism of down-regulation of p130 expression in proliferating cells was investigated. Our results indicate that the decline of p130 expression as G0 cells reenter the cell cycle is due to a decrease in protein stability. The enhancement of p130 turnover in late G1 and S phase compared with G0 and early G1 phase was dependent on Cdk4/6-specific phosphorylation of p130 on Serine 672, and independent of Cdk2 activity. The activity of the ubiquitin ligase complex Skp1-Cul1/Cdc53-F-box protein Skp2 (SCF(Skp2)) and the proteasome were necessary for p130 degradation. In vitro, recombinant Skp2 was able to bind hyperphosphorylated but not dephosphorylated p130. Furthermore, in vitro polyubiquitination of p130 by SCF(Skp2) was specifically dependent on phosphorylation of p130 on Serine 672. Thus, like the Cdk inhibitor p27(Kip1), p130 turnover is regulated by Cdk-dependent G1 phosphorylation leading to ubiquitin-dependent proteolysis.


CDC2-CDC28 Kinases , Cell Cycle Proteins , Ligases/metabolism , Phosphoproteins/metabolism , Proteins , Proto-Oncogene Proteins , Saccharomyces cerevisiae Proteins , Adaptor Proteins, Signal Transducing , Cell Cycle/physiology , Cells, Cultured , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinases/metabolism , Cysteine Endopeptidases/metabolism , Fungal Proteins/metabolism , Humans , Ligases/genetics , Multienzyme Complexes/metabolism , Phosphoproteins/genetics , Phosphorylation , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma-Like Protein p130 , Serine/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases
...