Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Syst Biol ; 17(6): e9522, 2021 06.
Article in English | MEDLINE | ID: mdl-34101353

ABSTRACT

Single-cell RNA sequencing has emerged as a powerful tool for resolving cellular states associated with normal and maligned developmental processes. Here, we used scRNA-seq to examine the cell cycle states of expanding human neural stem cells (hNSCs). From these data, we constructed a cell cycle classifier that identifies traditional cell cycle phases and a putative quiescent-like state in neuroepithelial-derived cell types during mammalian neurogenesis and in gliomas. The Neural G0 markers are enriched with quiescent NSC genes and other neurodevelopmental markers found in non-dividing neural progenitors. Putative glioblastoma stem-like cells were significantly enriched in the Neural G0 cell population. Neural G0 cell populations and gene expression are significantly associated with less aggressive tumors and extended patient survival for gliomas. Genetic screens to identify modulators of Neural G0 revealed that knockout of genes associated with the Hippo/Yap and p53 pathways diminished Neural G0 in vitro, resulting in faster G1 transit, down-regulation of quiescence-associated markers, and loss of Neural G0 gene expression. Thus, Neural G0 represents a dynamic quiescent-like state found in neuroepithelial-derived cells and gliomas.


Subject(s)
Glioblastoma , Neural Stem Cells , Animals , Cell Cycle/genetics , Cell Division , Humans , Neurogenesis/genetics
2.
Cancer Res ; 77(20): 5518-5529, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28855212

ABSTRACT

Glioblastoma multiforme (GBM) remains a mainly incurable disease in desperate need of more effective treatments. In this study, we develop evidence that the mitotic spindle checkpoint molecule BUB1B may offer a predictive marker for aggressiveness and effective drug response. A subset of GBM tumor isolates requires BUB1B to suppress lethal kinetochore-microtubule attachment defects. Using gene expression data from GBM stem-like cells, astrocytes, and neural progenitor cells that are sensitive or resistant to BUB1B inhibition, we created a computational framework to predict sensitivity to BUB1B inhibition. Applying this framework to tumor expression data from patients, we stratified tumors into BUB1B-sensitive (BUB1BS) or BUB1B-resistant (BUB1BR) subtypes. Through this effort, we found that BUB1BS patients have a significantly worse prognosis regardless of tumor development subtype (i.e., classical, mesenchymal, neural, proneural). Functional genomic profiling of BUB1BR versus BUB1BS isolates revealed a differential reliance of genes enriched in the BUB1BS classifier, including those involved in mitotic cell cycle, microtubule organization, and chromosome segregation. By comparing drug sensitivity profiles, we predicted BUB1BS cells to be more sensitive to type I and II topoisomerase inhibitors, Raf inhibitors, and other drugs, and experimentally validated some of these predictions. Taken together, the results show that our BUB1BR/S classification of GBM tumors can predict clinical course and sensitivity to drug treatment. Cancer Res; 77(20); 5518-29. ©2017 AACR.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Cell Cycle Proteins/antagonists & inhibitors , Glioblastoma/drug therapy , Glioblastoma/enzymology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor/metabolism , Etoposide/pharmacology , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Irinotecan , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics
3.
Oncotarget ; 8(30): 48545-48562, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28596487

ABSTRACT

Zinc finger domain genes comprise ~3% of the human genome, yet many of their functions remain unknown. Here we investigated roles for the vertebrate-specific BTB domain zinc finger gene ZNF131 in the context of human brain tumors. We report that ZNF131 is broadly required for Glioblastoma stem-like cell (GSC) viability, but dispensable for neural progenitor cell (NPC) viability. Examination of gene expression changes after ZNF131 knockdown (kd) revealed that ZNF131 activity notably promotes expression of Joubert Syndrome ciliopathy genes, including KIF7, NPHP1, and TMEM237, as well as HAUS5, a component of Augmin/HAUS complex that facilitates microtubule nucleation along the mitotic spindle. Of these genes only kd of HAUS5 displayed GSC-specific viability loss. Critically, HAUS5 ectopic expression was sufficient to suppress viability defects of ZNF131 kd cells. Moreover, ZNF131 and HAUS5 kd phenocopied each other in GSCs, each causing: mitotic arrest, centrosome fragmentation, loss of Augmin/HAUS complex on the mitotic spindle, and loss of GSC self-renewal and tumor formation capacity. In control NPCs, we observed centrosome fragmentation and lethality only when HAUS5 kd was combined with kd of HAUS2 or HAUS4, demonstrating that the complex is essential in NPCs, but that GSCs have heightened requirement. Our results suggest that GSCs differentially rely on ZNF131-dependent expression of HAUS5 as well as the Augmin/HAUS complex activity to maintain the integrity of centrosome function and viability.


Subject(s)
Brain Neoplasms/genetics , Centrosome/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , Transcription Factors/genetics , Brain Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Glioblastoma/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Binding , Spindle Apparatus/metabolism , Transcription Factors/metabolism
4.
Cell Syst ; 3(2): 172-186, 2016 08.
Article in English | MEDLINE | ID: mdl-27426982

ABSTRACT

We developed the transcription factor (TF)-target gene database and the Systems Genetics Network Analysis (SYGNAL) pipeline to decipher transcriptional regulatory networks from multi-omic and clinical patient data, and we applied these tools to 422 patients with glioblastoma multiforme (GBM). The resulting gbmSYGNAL network predicted 112 somatically mutated genes or pathways that act through 74 TFs and 37 microRNAs (miRNAs) (67 not previously associated with GBM) to dysregulate 237 distinct co-regulated gene modules associated with patient survival or oncogenic processes. The regulatory predictions were associated to cancer phenotypes using CRISPR-Cas9 and small RNA perturbation studies and also demonstrated GBM specificity. Two pairwise combinations (ETV6-NFKB1 and romidepsin-miR-486-3p) predicted by the gbmSYGNAL network had synergistic anti-proliferative effects. Finally, the network revealed that mutations in NF1 and PIK3CA modulate IRF1-mediated regulation of MHC class I antigen processing and presentation genes to increase tumor lymphocyte infiltration and worsen prognosis. Importantly, SYGNAL is widely applicable for integrating genomic and transcriptomic measurements from other human cohorts.


Subject(s)
Glioblastoma , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , MicroRNAs , Oncogenes
5.
Cell Rep ; 13(11): 2425-2439, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26673326

ABSTRACT

To identify therapeutic targets for glioblastoma (GBM), we performed genome-wide CRISPR-Cas9 knockout (KO) screens in patient-derived GBM stem-like cells (GSCs) and human neural stem/progenitors (NSCs), non-neoplastic stem cell controls, for genes required for their in vitro growth. Surprisingly, the vast majority GSC-lethal hits were found outside of molecular networks commonly altered in GBM and GSCs (e.g., oncogenic drivers). In vitro and in vivo validation of GSC-specific targets revealed several strong hits, including the wee1-like kinase, PKMYT1/Myt1. Mechanistic studies demonstrated that PKMYT1 acts redundantly with WEE1 to inhibit cyclin B-CDK1 activity via CDK1-Y15 phosphorylation and to promote timely completion of mitosis in NSCs. However, in GSCs, this redundancy is lost, most likely as a result of oncogenic signaling, causing GBM-specific lethality.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Cycle Proteins/genetics , Genome, Human , Membrane Proteins/genetics , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Cyclin B/metabolism , ErbB Receptors/metabolism , Gene Library , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Microscopy, Video , Mitosis , Neoplastic Stem Cells/cytology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , RNA Interference , Time-Lapse Imaging , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Clin Cancer Res ; 21(2): 233-9, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25104085

ABSTRACT

Kinetochores are large protein structures assembled on centromeric DNA during mitosis that bind to microtubules of the mitotic spindle to orchestrate and power chromosome movements. Deregulation of kinetochore-microtubule (KT-MT) attachments has been implicated in driving chromosome instability and cancer evolution; however, the nature and source of KT-MT attachment defects in cancer cells remain largely unknown. Here, we highlight recent findings suggesting that oncogene-driven changes in kinetochore regulation occur in glioblastoma multiforme (GBM) and possibly other cancers exhibiting chromosome instability, giving rise to novel therapeutic opportunities. In particular, we consider the GLE2p-binding sequence domains of BubR1 and the newly discovered BuGZ, two kinetochore-associated proteins, as candidate therapeutic targets for GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Kinetochores/physiology , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Humans , Kinetochores/drug effects , M Phase Cell Cycle Checkpoints , Microtubules/drug effects , Molecular Targeted Therapy , Neoplasms/pathology , Protein Binding
7.
Dev Cell ; 28(3): 282-94, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24462187

ABSTRACT

During mitosis, the spindle assembly checkpoint (SAC) monitors the attachment of kinetochores (KTs) to the plus ends of spindle microtubules (MTs) and prevents anaphase onset until chromosomes are aligned and KTs are under proper tension. Here, we identify a SAC component, BuGZ/ZNF207, from an RNAi viability screen in human glioblastoma multiforme (GBM) brain tumor stem cells. BuGZ binds to and stabilizes Bub3 during interphase and mitosis through a highly conserved GLE2p-binding sequence (GLEBS) domain. Inhibition of BuGZ results in loss of both Bub3 and its binding partner Bub1 from KTs, reduction of Bub1-dependent phosphorylation of centromeric histone H2A, attenuation of KT-based Aurora B kinase activity, and lethal chromosome congression defects in cancer cells. Phylogenetic analysis indicates that BuGZ orthologs are highly conserved among eukaryotes, but are conspicuously absent from budding and fission yeasts. These findings suggest that BuGZ has evolved to facilitate Bub3 activity and chromosome congression in higher eukaryotes.


Subject(s)
Cell Cycle Proteins/chemistry , Chromosomes, Human/genetics , Glioblastoma/pathology , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/physiology , Zinc Fingers/genetics , Amino Acid Sequence , Animals , Aurora Kinase B/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Fluorescent Antibody Technique , Glioblastoma/genetics , Glioblastoma/metabolism , Histones/metabolism , Humans , Immunoblotting , Mice , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Mitosis/physiology , Molecular Sequence Data , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation , Phylogeny , Poly-ADP-Ribose Binding Proteins , Protein Stability , RNA, Small Interfering/genetics , Sequence Homology, Amino Acid
8.
Genes Dev ; 27(9): 1032-45, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23651857

ABSTRACT

To identify key regulators of human brain tumor maintenance and initiation, we performed multiple genome-wide RNAi screens in patient-derived glioblastoma multiforme (GBM) stem cells (GSCs). These screens identified the plant homeodomain (PHD)-finger domain protein PHF5A as differentially required for GSC expansion, as compared with untransformed neural stem cells (NSCs) and fibroblasts. Given PHF5A's known involvement in facilitating interactions between the U2 snRNP complex and ATP-dependent helicases, we examined cancer-specific roles in RNA splicing. We found that in GSCs, but not untransformed controls, PHF5A facilitates recognition of exons with unusual C-rich 3' splice sites in thousands of essential genes. PHF5A knockdown in GSCs, but not untransformed NSCs, astrocytes, or fibroblasts, inhibited splicing of these genes, leading to cell cycle arrest and loss of viability. Notably, pharmacologic inhibition of U2 snRNP activity phenocopied PHF5A knockdown in GSCs and also in NSCs or fibroblasts overexpressing MYC. Furthermore, PHF5A inhibition compromised GSC tumor formation in vivo and inhibited growth of established GBM patient-derived xenograft tumors. Our results demonstrate a novel viability requirement for PHF5A to maintain proper exon recognition in brain tumor-initiating cells and may provide new inroads for novel anti-GBM therapeutic strategies.


Subject(s)
Brain Neoplasms/physiopathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Glioblastoma/physiopathology , RNA Interference , Animals , Brain Neoplasms/genetics , Cell Cycle Checkpoints , Cell Line , Cell Proliferation , Cell Survival/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Glioblastoma/genetics , Humans , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Splicing , RNA-Binding Proteins , Trans-Activators , Transplantation, Heterologous
9.
Cancer Discov ; 3(2): 198-211, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23154965

ABSTRACT

UNLABELLED: To identify new candidate therapeutic targets for glioblastoma multiforme, we combined functional genetics and glioblastoma network modeling to identify kinases required for the growth of patient-derived brain tumor-initiating cells (BTIC) but that are dispensable to proliferating human neural stem cells (NSC). This approach yielded BUB1B/BUBR1, a critical mitotic spindle checkpoint player, as the top-scoring glioblastoma lethal kinase. Knockdown of BUB1B inhibited expansion of BTIC isolates, both in vitro and in vivo, without affecting proliferation of NSCs or astrocytes. Mechanistic studies revealed that BUB1B's GLE2p-binding sequence (GLEBS) domain activity is required to suppress lethal kinetochore-microtubule (KT-MT) attachment defects in glioblastoma isolates and genetically transformed cells with altered sister KT dynamics, which likely favor KT-MT instability. These results indicate that glioblastoma tumors have an added requirement for BUB1B to suppress lethal consequences of altered KT function and further suggest that sister KT measurements may predict cancer-specific sensitivity to BUB1B inhibition and perhaps other mitotic targets that affect KT-MT stability. SIGNIFICANCE: Currently, no effective therapies are available for glioblastoma, the most frequent and aggressive brain tumor. Our results suggest that targeting the GLEBS domain activity of BUB1B may provide a therapeutic window for glioblastoma, as the GLEBS domain is nonessential in untransformed cells. Moreover, the results further suggest that sister KT distances at metaphase may predict sensitivity to anticancer therapeutics targeting KT function.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Binding Sites/genetics , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle Proteins , Cell Line , Cell Line, Transformed , Cell Proliferation , Genetic Predisposition to Disease/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , HeLa Cells , Humans , Kinetochores/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplastic Stem Cells/pathology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Spindle Apparatus/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
10.
J Immunol ; 186(11): 6576-84, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21525393

ABSTRACT

One pathological hallmark of HIV-1 infection is chronic activation of the immune system, driven, in part, by increased expression of proinflammatory cytokines. The host attempts to counterbalance this prolonged immune activation through compensatory mediators of immune suppression. We recently identified a gene encoding the proinflammatory cytokine IL-32 in microarray studies of HIV-1 infection in lymphatic tissue (LT) and show in this study that increased expression of IL-32 in both gut and LT of HIV-1-infected individuals may have a heretofore unappreciated role as a mediator of immune suppression. We show that: 1) IL-32 expression is increased in CD4(+) T cells, B cells, macrophages, dendritic cells, and epithelial cells in vivo; 2) IL-32 induces the expression of immunosuppressive molecules IDO and Ig-like transcript 4 in immune cells in vitro; and 3) in vivo, IL-32-associated IDO/Ig-like transcript 4 expression in LT macrophages and gut epithelial cells decreases immune activation but also may impair host defenses, supporting productive viral replication, thereby accounting for the correlation between IL-32 levels and HIV-1 replication in LT. Thus, during HIV-1 infection, we propose that IL-32 moderates chronic immune activation to avert associated immunopathology but at the same time dampens the antiviral immune response and thus paradoxically supports HIV-1 replication and viral persistence.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Interleukins/immunology , Lymphoid Tissue/immunology , Adult , Female , Gene Expression Profiling , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Host-Pathogen Interactions/immunology , Humans , Ileum/immunology , Ileum/metabolism , Ileum/virology , In Situ Hybridization , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukins/genetics , Interleukins/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/virology , Lymphoid Tissue/metabolism , Lymphoid Tissue/virology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Viral/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Rectum/immunology , Rectum/metabolism , Rectum/virology , Time Factors , Virus Replication/genetics , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...