Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell ; 157(2): 447-458, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725410

ABSTRACT

Maintenance of a constant cell volume in response to extracellular or intracellular osmotic changes is critical for cellular homeostasis. Activation of a ubiquitous volume-regulated anion channel (VRAC) plays a key role in this process; however, its molecular identity in vertebrates remains unknown. Here, we used a cell-based fluorescence assay and performed a genome-wide RNAi screen to find components of VRAC. We identified SWELL1 (LRRC8A), a member of a four-transmembrane protein family with unknown function, as essential for hypotonicity-induced iodide influx. SWELL1 is localized to the plasma membrane, and its knockdown dramatically reduces endogenous VRAC currents and regulatory cell volume decrease in various cell types. Furthermore, point mutations in SWELL1 cause a significant change in VRAC anion selectivity, demonstrating that SWELL1 is an essential VRAC component. These findings enable further molecular characterization of the VRAC channel complex and genetic studies for understanding the function of VRAC in normal physiology and disease.


Subject(s)
Cell Size , Membrane Proteins/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Genome-Wide Association Study , HEK293 Cells , HeLa Cells , Humans , Iodides/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , RNA Interference
2.
Cell ; 139(1): 199-210, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19765810

ABSTRACT

Two decades of research identified more than a dozen clock genes and defined a biochemical feedback mechanism of circadian oscillator function. To identify additional clock genes and modifiers, we conducted a genome-wide small interfering RNA screen in a human cellular clock model. Knockdown of nearly 1000 genes reduced rhythm amplitude. Potent effects on period length or increased amplitude were less frequent; we found hundreds of these and confirmed them in secondary screens. Characterization of a subset of these genes demonstrated a dosage-dependent effect on oscillator function. Protein interaction network analysis showed that dozens of gene products directly or indirectly associate with known clock components. Pathway analysis revealed these genes are overrepresented for components of insulin and hedgehog signaling, the cell cycle, and the folate metabolism. Coupled with data showing many of these pathways are clock regulated, we conclude the clock is interconnected with many aspects of cellular function.


Subject(s)
Biological Clocks , Circadian Rhythm , Genome-Wide Association Study , Cell Line , Gene Knockdown Techniques , Humans , RNA Interference , RNA, Small Interfering/metabolism
3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443164

ABSTRACT

The regulatory mechanisms of circadian rhythms have been studied primarily at the level of the transcription-translation feedback loops of protein-coding genes. Regulatory modules involving noncoding RNAs are less thoroughly understood. In particular, emerging evidence has revealed the important role of microRNAs (miRNAs) in maintaining the robustness of the circadian system. To identify miRNAs that have the potential to modulate circadian rhythms, we conducted a genome-wide miRNA screen using U2OS luciferase reporter cells. Among 989 miRNAs in the library, 120 changed the period length in a dose-dependent manner. We further validated the circadian regulatory function of an miRNA cluster, miR-183/96/182, both in vitro and in vivo. We found that all three members of this miRNA cluster can modulate circadian rhythms. Particularly, miR-96 directly targeted a core circadian clock gene, PER2. The knockout of the miR-183/96/182 cluster in mice showed tissue-specific effects on circadian parameters and altered circadian rhythms at the behavioral level. This study identified a large number of miRNAs, including the miR-183/96/182 cluster, as circadian modulators. We provide a resource for further understanding the role of miRNAs in the circadian network and highlight the importance of miRNAs as a genome-wide layer of circadian clock regulation.


Subject(s)
Circadian Rhythm/genetics , Gene Expression Regulation/genetics , MicroRNAs/metabolism , Period Circadian Proteins/metabolism , Animals , Cell Line, Tumor , Circadian Rhythm/radiation effects , Gene Expression Regulation/radiation effects , Gene Knock-In Techniques , Gene Knockout Techniques , Genomics , Humans , Luciferases/genetics , Luciferases/metabolism , Lung/metabolism , Lung/radiation effects , Mice , MicroRNAs/genetics , Multigene Family , Organ Specificity , Period Circadian Proteins/genetics , Retina/metabolism , Retina/radiation effects , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/radiation effects , Time Factors
4.
Cell ; 135(1): 49-60, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18854154

ABSTRACT

Human Immunodeficiency Viruses (HIV-1 and HIV-2) rely upon host-encoded proteins to facilitate their replication. Here, we combined genome-wide siRNA analyses with interrogation of human interactome databases to assemble a host-pathogen biochemical network containing 213 confirmed host cellular factors and 11 HIV-1-encoded proteins. Protein complexes that regulate ubiquitin conjugation, proteolysis, DNA-damage response, and RNA splicing were identified as important modulators of early-stage HIV-1 infection. Additionally, over 40 new factors were shown to specifically influence the initiation and/or kinetics of HIV-1 DNA synthesis, including cytoskeletal regulatory proteins, modulators of posttranslational modification, and nucleic acid-binding proteins. Finally, 15 proteins with diverse functional roles, including nuclear transport, prostaglandin synthesis, ubiquitination, and transcription, were found to influence nuclear import or viral DNA integration. Taken together, the multiscale approach described here has uncovered multiprotein virus-host interactions that likely act in concert to facilitate the early steps of HIV-1 infection.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Proteins/metabolism , Virus Replication , Cell Line , Humans , RNA Interference , Two-Hybrid System Techniques
5.
Nature ; 541(7638): 541-545, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28068668

ABSTRACT

Cell fate perturbations underlie many human diseases, including breast cancer. Unfortunately, the mechanisms by which breast cell fate are regulated are largely unknown. The mammary gland epithelium consists of differentiated luminal epithelial and basal myoepithelial cells, as well as undifferentiated stem cells and more restricted progenitors. Breast cancer originates from this epithelium, but the molecular mechanisms that underlie breast epithelial hierarchy remain ill-defined. Here, we use a high-content confocal image-based short hairpin RNA screen to identify tumour suppressors that regulate breast cell fate in primary human breast epithelial cells. We show that ablation of the large tumour suppressor kinases (LATS) 1 and 2 (refs 5, 6), which are part of the Hippo pathway, promotes the luminal phenotype and increases the number of bipotent and luminal progenitors, the proposed cells-of-origin of most human breast cancers. Mechanistically, we have identified a direct interaction between Hippo and oestrogen receptor-α (ERα) signalling. In the presence of LATS, ERα was targeted for ubiquitination and Ddb1-cullin4-associated-factor 1 (DCAF1)-dependent proteasomal degradation. Absence of LATS stabilized ERα and the Hippo effectors YAP and TAZ (hereafter YAP/TAZ), which together control breast cell fate through intrinsic and paracrine mechanisms. Our findings reveal a non-canonical (that is, YAP/TAZ-independent) effect of LATS in the regulation of human breast cell fate.


Subject(s)
Breast/cytology , Breast/enzymology , Cell Differentiation , Cell Lineage , Estrogen Receptor alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/agonists , Adaptor Proteins, Signal Transducing/metabolism , Breast/pathology , Carrier Proteins/metabolism , Cells, Cultured , Estrogen Receptor alpha/agonists , Female , Genes, Tumor Suppressor , Humans , Phosphoproteins/agonists , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/deficiency , Proteolysis , Signal Transduction , Transcription Factors , Tumor Suppressor Proteins/deficiency , Ubiquitin/metabolism , Ubiquitin-Protein Ligases , YAP-Signaling Proteins
6.
Nat Methods ; 15(11): 941-946, 2018 11.
Article in English | MEDLINE | ID: mdl-30297964

ABSTRACT

CRISPR-Cas9 screening allows genome-wide interrogation of gene function. Currently, to achieve the high and uniform Cas9 expression desirable for screening, one needs to engineer stable and clonal Cas9-expressing cells-an approach that is not applicable in human primary cells. Guide Swap permits genome-scale pooled CRISPR-Cas9 screening in human primary cells by exploiting the unexpected finding that editing by lentivirally delivered, targeted guide RNAs (gRNAs) occurs efficiently when Cas9 is introduced in complex with nontargeting gRNA. We validated Guide Swap in depletion and enrichment screens in CD4+ T cells. Next, we implemented Guide Swap in a model of ex vivo hematopoiesis, and identified known and previously unknown regulators of CD34+ hematopoietic stem and progenitor cell (HSPC) expansion. We anticipate that this platform will be broadly applicable to other challenging cell types, and thus will enable discovery in previously inaccessible but biologically relevant human primary cell systems.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Gene Editing , Genome, Human , Hematopoietic Stem Cells/metabolism , RNA, Guide, Kinetoplastida/genetics , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans
7.
Mol Cell ; 50(3): 356-67, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23603119

ABSTRACT

A high-throughput RNA interference (RNAi) screen targeting 542 genes of the human kinome was used to discover regulators of RNAi. Here we report that the proto-oncogene Akt-3/PKBγ (Akt3) phosphorylates Argonaute 2 (Ago2) at S387, which downregulates cleavage and upregulates translational repression of endogenous microRNA (miRNA)-targeted messenger RNAs (mRNAs). We further demonstrate that Akt3 coimmunoprecipitates with Ago2 and phosphorylation of Ago2 at S387 facilitates its interaction with GW182 and localization to cytoplasmic processing bodies (P bodies), where miRNA-targeted mRNAs are thought to be stored and degraded. Therefore, Akt3-mediated phosphorylation of Ago2 is a molecular switch between target mRNA cleavage and translational repression activities of Ago2.


Subject(s)
Argonaute Proteins/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , Argonaute Proteins/metabolism , Cell Line , Cell Line, Tumor , Down-Regulation , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation
8.
Nat Genet ; 38(3): 312-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16474406

ABSTRACT

Direct evidence for the requirement of transcriptional feedback repression in circadian clock function has been elusive. Here, we developed a molecular genetic screen in mammalian cells to identify mutants of the circadian transcriptional activators CLOCK and BMAL1, which were uncoupled from CRYPTOCHROME (CRY)-mediated transcriptional repression. Notably, mutations in the PER-ARNT-SIM domain of CLOCK and the C terminus of BMAL1 resulted in synergistic insensitivity through reduced physical interactions with CRY. Coexpression of these mutant proteins in cultured fibroblasts caused arrhythmic phenotypes in population and single-cell assays. These data demonstrate that CRY-mediated repression of the CLOCK/BMAL1 complex activity is required for maintenance of circadian rhythmicity and provide formal proof that transcriptional feedback is required for mammalian clock function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Circadian Rhythm/physiology , Gene Expression Regulation , Trans-Activators/genetics , 3T3 Cells , ARNTL Transcription Factors , Animals , CLOCK Proteins , Cell Line , Feedback , Genes, Reporter , Humans , Luciferases/analysis , Luciferases/genetics , Luminescence , Mice , Plasmids , Time
9.
Proc Natl Acad Sci U S A ; 108(41): 17117-22, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21949408

ABSTRACT

Glucocorticoids can inhibit inflammation by abrogating the activity of NF-κB, a family of transcription factors that regulates the production of proinflammatory cytokines. To understand the molecular mechanism of repression of NF-κB activity by glucocorticoids, we performed a high-throughput siRNA oligo screen to identify novel genes involved in this process. Here, we report that loss of p53, a tumor suppressor protein, impaired repression of NF-κB target gene transcription by glucocorticoids. Additionally, loss of p53 also impaired transcription of glucocorticoid receptor (GR) target genes, whereas upstream NF-κB and glucocorticoid receptor signaling cascades remained intact. We further demonstrate that p53 loss severely impaired glucocorticoid rescue of death in a mouse model of LPS shock. Our findings unveil a new role for p53 in the repression of NF-κB by glucocorticoids and suggest important implications for treatment of the proinflammatory microenvironments found in tumors with aberrant p53 activity.


Subject(s)
NF-kappa B/metabolism , Receptors, Glucocorticoid/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Dexamethasone/pharmacology , High-Throughput Screening Assays , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Interaction Maps , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/metabolism , Signal Transduction , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
10.
Proc Natl Acad Sci U S A ; 104(51): 20314-9, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18077367

ABSTRACT

Signal transduction pathways often use a transcriptional component to mediate adaptive cellular responses. Coactivator proteins function prominently in these pathways as the conduit to the basic transcriptional machinery. Here we present a high-throughput cell-based screening strategy, termed the "coactivator trap," to study the functional interactions of coactivators with transcription factors. We applied this strategy to the cAMP signaling pathway, which utilizes two families of coactivators, the cAMP response element binding protein (CREB) binding protein (CBP)/p300 family and the recently identified transducers of regulated CREB activity family (TORCs1-3). In addition to identifying numerous known interactions of these coactivators, this analysis identified NONO (p54(nrb)) as a TORC-interacting protein. RNA interference experiments demonstrate that NONO is necessary for cAMP-dependent activation of CREB target genes in vivo. Furthermore, TORC2 and NONO complex on cAMP-responsive promoters, and NONO acts as a bridge between the CREB/TORC complex and RNA polymerase II. These data demonstrate the utility of the coactivator trap by identification of a component of cAMP-mediated transcription.


Subject(s)
Cyclic AMP/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Octamer Transcription Factors/metabolism , Protein Interaction Mapping/methods , RNA-Binding Proteins/metabolism , Cell Line , DNA-Binding Proteins , Humans , Nuclear Matrix-Associated Proteins/antagonists & inhibitors , Nuclear Matrix-Associated Proteins/genetics , Octamer Transcription Factors/antagonists & inhibitors , Octamer Transcription Factors/genetics , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
11.
Neuron ; 43(4): 527-37, 2004 Aug 19.
Article in English | MEDLINE | ID: mdl-15312651

ABSTRACT

The mammalian circadian clock plays an integral role in timing rhythmic physiology and behavior, such as locomotor activity, with anticipated daily environmental changes. The master oscillator resides within the suprachiasmatic nucleus (SCN), which can maintain circadian rhythms in the absence of synchronizing light input. Here, we describe a genomics-based approach to identify circadian activators of Bmal1, itself a key transcriptional activator that is necessary for core oscillator function. Using cell-based functional assays, as well as behavioral and molecular analyses, we identified Rora as an activator of Bmal1 transcription within the SCN. Rora is required for normal Bmal1 expression and consolidation of daily locomotor activity and is regulated by the core clock in the SCN. These results suggest that opposing activities of the orphan nuclear receptors Rora and Rev-erb alpha, which represses Bmal1 expression, are important in the maintenance of circadian clock function.


Subject(s)
Biological Clocks/genetics , Circadian Rhythm/genetics , Genomics/methods , Receptors, Retinoic Acid/genetics , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Nuclear Receptor Subfamily 1, Group F, Member 1 , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear , Receptors, Retinoic Acid/biosynthesis , Response Elements/genetics , Trans-Activators , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Mol Cancer Ther ; 2(3): 273-81, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12657722

ABSTRACT

Protein kinase C (PKC) promotes cell survival in response to ionizing radiation in a variety of experimental models including human carcinoma, human glioblastoma, and transformed mouse embryo fibroblast cell lines. We have introduced specific antisense oligonucleotides into human mammary tumor cell lines in vitro to analyze the role of individual PKC isoforms in radiation-induced cell death in breast cancer. MDA-MB-231 and MCF-7 cells treated with oligonucleotide directed against the PKC delta isoform exhibited impaired survival in response to 5.6 Gy gamma-radiation as measured by mitochondrial metabolism of tetrazolium dye. The role of PKC delta in the breast tumor cell lines was of particular interest, because contradictory reports exist in the literature regarding the role of PKC delta in cell survival and apoptosis. A comparison of the effects of the PKC delta antisense oligonucleotide and a nucleotide scrambled version of this nucleotide revealed only the antisense oligonucleotide decreased cell survival. The PKC delta antisense oligonucleotide decreased cell survival after exposure to low (1.5 Gy) radiation doses and in the absence of radiation insult. We found 3 micro M rottlerin, a selective PKC delta inhibitor, to reduce MCF-7 and MDA-MB-231 cell survival. Furthermore, MCF-7 cells transformed to express a dominant-negative mutant of PKC delta exhibited reduced survival. Comet analysis showed that PKC delta oligonucleotide treatment caused an accumulation of cells containing damaged DNA similar to that seen in 1.5 Gy radiation-treated cells. We conclude that PKC delta acts as a prosurvival factor in human breast tumor cells in vitro.


Subject(s)
Breast Neoplasms/metabolism , Protein Kinase C/physiology , Acetophenones/pharmacology , Benzopyrans/pharmacology , Blotting, Western , Breast Neoplasms/pathology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Survival/physiology , Comet Assay , DNA, Neoplasm/genetics , Down-Regulation , Enzyme Inhibitors/pharmacology , Gamma Rays , Genes, Dominant , Humans , Isoenzymes , Oligoribonucleotides, Antisense/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
13.
Cell Host Microbe ; 11(3): 306-18, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22423970

ABSTRACT

Pathogens commonly utilize endocytic pathways to gain cellular access. The endosomal pattern recognition receptors TLR7 and TLR9 detect pathogen-encoded nucleic acids to initiate MyD88-dependent proinflammatory responses to microbial infection. Using genome-wide RNAi screening and integrative systems-based analysis, we identify 190 cofactors required for TLR7- and TLR9-directed signaling responses. A set of cofactors were crossprofiled for their activities downstream of several immunoreceptors and then functionally mapped based on the known architecture of NF-κB signaling pathways. Protein complexes and pathways involved in ubiquitin-protein ligase activities, sphingolipid metabolism, chromatin modifications, and ancient stress responses were found to modulate innate recognition of endosomal nucleic acids. Additionally, hepatocyte growth factor-regulated tyrosine kinase substrate (HRS) was characterized as necessary for ubiquitin-dependent TLR9 targeting to the endolysosome. Proteins and pathways identified here should prove useful in delineating strategies to manipulate innate responses for treatment of autoimmune disorders and microbial infection.


Subject(s)
Immunity, Innate/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Animals , Chick Embryo , Computer Simulation , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/physiology , Endosomes/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Gene Regulatory Networks , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Protein Transport , RNA Interference , Signal Transduction , Support Vector Machine
14.
Comb Chem High Throughput Screen ; 14(8): 648-57, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21564017

ABSTRACT

The luminescent reporter gene assay (LRGA) is arguably the most prominent type of reporter gene assay used in biomolecular and pharmaceutical development laboratories. Part of this popularity is due to the high signal associated with luciferases, the foundation of this technology. This feature makes them ideally suited for high throughput screening applications where potentially millions of chemical compounds can be analyzed in a given assay. Recent technical advancements that enhance signal stability of the luciferases along with development and commercialization of multiple forms of luciferases, their respective substrates, and improvements in expression vectors for reporter gene assay (RGA) applications have broadened their use. While the practical challenges related to the application of luminescent technology in a laboratory setting have been overcome, there remains much to do in laying a systematic approach towards the construction of RGAs, which are essential to the elucidation of the basic biology for genes of interest. This mini-review aims at giving a birds-eye view of the available luciferases, substrates and other luminescent technologies available and provides a general blueprint as well as practical considerations for constructing and interfacing RGAs with chemical biology and functional genomics for the elucidation of fundamental biological questions and for biomedical research.


Subject(s)
Genes, Reporter , Light , Luminescence
15.
Cell Stem Cell ; 6(1): 37-47, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-20085741

ABSTRACT

Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer associated with a very poor prognosis. Recently, the initiation and growth of GBM has been linked to brain tumor-initiating cells (BTICs), which are poorly differentiated and share features with neural stem cells (NSCs). Here we describe a kinome-wide RNA interference screen to identify factors that control the tumorigenicity of BTICs. We identified several genes whose silencing induces differentiation of BTICs derived from multiple GBM patients. In particular, knockdown of the adaptor protein TRRAP significantly increased differentiation of cultured BTICs, sensitized the cells to apoptotic stimuli, and negatively affected cell cycle progression. TRRAP knockdown also significantly suppressed tumor formation upon intracranial BTIC implantation into mice. Together, these findings support a critical role for TRRAP in maintaining a tumorigenic, stem cell-like state.


Subject(s)
Adaptor Proteins, Signal Transducing/analysis , Brain Neoplasms/chemistry , Cell Differentiation , Cell Transformation, Neoplastic/chemistry , Glioblastoma/chemistry , Neoplastic Stem Cells/chemistry , Nuclear Proteins/analysis , RNA Interference , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Mice, SCID , Neoplastic Stem Cells/cytology , Nuclear Proteins/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Proc Natl Acad Sci U S A ; 104(43): 16940-5, 2007 Oct 23.
Article in English | MEDLINE | ID: mdl-17939994

ABSTRACT

IkappaB kinase 2 (IKK2 or IKKbeta) is a component of the IKK complex that coordinates the cellular response to a diverse set of extracellular stimuli, including cytokines, microbial infection, and stress. In response to an external stimulus, the complex is activated, resulting in the phosphorylation and subsequent proteasome-mediated degradation of IkappaB proteins. This event triggers the nuclear import of the NF-kappaB transcription factor, which activates the transcription of genes that regulate a variety of fundamental biological processes, including immune response, cell survival, and development. Here, we define an essential role for IKK2 in normal mitotic progression and the maintenance of spindle bipolarity. Chemical and genetic perturbation of IKK2 promotes the formation of multipolar spindles and chromosome missegregation. Depletion of IKK2 results in the deregulation of Aurora A protein stability and coincident hyperactivation of a putative Aurora A substrate, the mitotic motor KIF11. These data support a function for IKK2 as an antagonist of Aurora A signaling during mitosis. Additionally, our results indicate a direct role for IKK2 in the maintenance of genome stability and underscore the potential for oncogenic consequences in targeting this kinase for therapeutic intervention.


Subject(s)
I-kappa B Kinase/metabolism , Spindle Apparatus/enzymology , Aneuploidy , Animals , Aurora Kinase A , Aurora Kinases , Cell Cycle/drug effects , Cell Polarity/drug effects , Cell Transformation, Neoplastic/drug effects , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , HeLa Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/deficiency , Mice , Mice, Nude , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Spindle Apparatus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL