Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-37609141

ABSTRACT

Cancer cells are often aneuploid and frequently display elevated rates of chromosome missegregation in a phenomenon called chromosomal instability (CIN). CIN is commonly caused by hyperstable kinetochore-microtubule (K-MT) attachments that reduces the efficiency of correction of erroneous K-MT attachments. We recently showed that UMK57, a chemical agonist of MCAK (alias KIF2C) improves chromosome segregation fidelity in CIN cancer cells although cells rapidly develop adaptive resistance. To determine the mechanism of resistance we performed unbiased proteomic screens which revealed increased phosphorylation in cells adapted to UMK57 at two Aurora kinase A phosphoacceptor sites on BOD1L1 (alias FAM44A). BOD1L1 depletion or Aurora kinase A inhibition eliminated resistance to UMK57 in CIN cancer cells. BOD1L1 localizes to spindles/kinetochores during mitosis, interacts with the PP2A phosphatase, and regulates phosphorylation levels of kinetochore proteins, chromosome alignment, mitotic progression and fidelity. Moreover, the BOD1L1 gene is mutated in a subset of human cancers, and BOD1L1 depletion reduces cell growth in combination with clinically relevant doses of taxol or Aurora kinase A inhibitor. Thus, an Aurora kinase A -BOD1L1-PP2A axis promotes faithful chromosome segregation during mitosis.

2.
Stem Cell Reports ; 18(2): 475-488, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36638786

ABSTRACT

During in vitro propagation, human pluripotent stem cells (hPSCs) frequently become aneuploid with incorrect chromosome numbers due to mitotic chromosome segregation errors. Yet, it is not understood why hPSCs exhibit a low mitotic fidelity. Here, we investigate the mechanisms responsible for mitotic errors in hPSCs and show that the primary cause is lagging chromosomes in anaphase with improper merotelic microtubule attachments. Accordingly, short-term treatment (<24 h) with small molecules that prolong mitotic duration or destabilize chromosome microtubule attachments reduces merotelic errors and lagging chromosome rates, although hPSCs adapt and lagging chromosome rates rebound upon long-term (>24 h) microtubule destabilization. Strikingly, we also demonstrate that mitotic error rates correlate with developmental potential decreasing or increasing upon loss or gain of pluripotency, respectively. Thus, a low mitotic fidelity is an inherent and conserved phenotype of hPSCs. Moreover, chromosome segregation fidelity depends on developmental state in normal human cells.


Subject(s)
Chromosome Segregation , Kinetochores , Humans , Mitosis , Microtubules , Anaphase , Spindle Apparatus
3.
bioRxiv ; 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38187612

ABSTRACT

To ensure genomic fidelity a series of spatially and temporally coordinated events are executed during prometaphase of mitosis, including bipolar spindle formation, chromosome attachment to spindle microtubules at kinetochores, the correction of erroneous kinetochore-microtubule (k-MT) attachments, and chromosome congression to the spindle equator. Cyclin A/Cdk1 kinase plays a key role in destabilizing k-MT attachments during prometaphase to promote correction of erroneous k-MT attachments. However, it is unknown if Cyclin A/Cdk1 kinase regulates other events during prometaphase. Here, we investigate additional roles of Cyclin A/Cdk1 in prometaphase by using an siRNA knockdown strategy to deplete endogenous Cyclin A from human cells. We find that depleting Cyclin A significantly extends mitotic duration, specifically prometaphase, because chromosome alignment is delayed. Unaligned chromosomes display erroneous monotelic, syntelic, or lateral k-MT attachments suggesting that bioriented k-MT attachment formation is delayed in the absence of Cyclin A. Mechanistically, chromosome alignment is likely impaired because the localization of the kinetochore proteins BUB1 kinase, KNL1, and MPS1 kinase are reduced in Cyclin A-depleted cells. Moreover, we find that Cyclin A promotes BUB1 kinetochore localization independently of its role in destabilizing k-MT attachments. Thus, Cyclin A/Cdk1 facilitates chromosome alignment during prometaphase to support timely mitotic progression.

4.
J Cell Biol ; 221(9)2022 09 05.
Article in English | MEDLINE | ID: mdl-35878017

ABSTRACT

Kinetochore protein phosphorylation promotes the correction of erroneous microtubule attachments to ensure faithful chromosome segregation during cell division. Determining how phosphorylation executes error correction requires an understanding of whether kinetochore substrates are completely (i.e., all-or-none) or only fractionally phosphorylated. Using quantitative mass spectrometry (MS), we measured phospho-occupancy on the conserved kinetochore protein Hec1 (NDC80) that directly binds microtubules. None of the positions measured exceeded ∼50% phospho-occupancy, and the cumulative phospho-occupancy changed by only ∼20% in response to changes in microtubule attachment status. The narrow dynamic range of phospho-occupancy is maintained, in part, by the ongoing phosphatase activity. Further, both Cdk1-Cyclin B1 and Aurora kinases phosphorylate Hec1 to enhance error correction in response to different types of microtubule attachment errors. The low inherent phospho-occupancy promotes microtubule attachment to kinetochores while the high sensitivity of kinetochore-microtubule attachments to small changes in phospho-occupancy drives error correction and ensures high mitotic fidelity.


Subject(s)
Cytoskeletal Proteins , Kinetochores , Microtubules , Mitosis , Aurora Kinases/metabolism , CDC2 Protein Kinase/metabolism , Chromosome Segregation , Cyclin B1/metabolism , Cytoskeletal Proteins/metabolism , HeLa Cells , Humans , Kinetochores/metabolism , Microtubules/metabolism , Phosphorylation
5.
Methods Mol Biol ; 2415: 175-182, 2022.
Article in English | MEDLINE | ID: mdl-34972954

ABSTRACT

Cyclin A promotes Cdk activity in a cell cycle-dependent manner to facilitate specific cell cycle events and transitions with an established role for DNA replication in S phase. Recent evidence demonstrates that cyclin A also activates Cdk during early mitosis to promote faithful chromosome segregation by regulating the stability of kinetochore-microtubule (k-MT) attachments. Here we describe a methodology to identify protein substrates of cyclin A/Cdk during mitosis in human cells. The method combines selective cell cycle synchrony in mitosis with stable isotope labeling of amino acids in cell culture (SILAC) coupled to mass spectrometry. This strategy identified a catalogue of potential cyclin A/Cdk substrates in mitosis, as well as unveiled potential intersections between signaling regulated by Aurora, Polo-like, and Cdk mitotic kinases.


Subject(s)
Cyclin A , Mitosis , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation , Cyclin A/genetics , Cyclin A/metabolism , Humans , Microtubules/metabolism , Phosphorylation
6.
Methods Cell Biol ; 158: 91-116, 2020.
Article in English | MEDLINE | ID: mdl-32423652

ABSTRACT

During mitosis, spindle microtubules dynamically attach to and detach from kinetochores in a precise and regulated fashion. To ensure mitotic fidelity, kinetochore-microtubule (k-MT) attachments must be stable enough to satisfy the spindle assembly checkpoint (SAC), but sufficiently unstable to facilitate the correction of maloriented attachments. Different methods are available to assess k-MT stability in both live and fixed cells, but a comparative survey of these methods has not yet been reported. Here, we evaluate several quantitative and semiquantitative methods for determining k-MT stability and apply each technique to illustrate changes in spindle microtubule dynamics upon perturbation with physiologically relevant concentrations of microtubule stabilizing (Taxol) and destabilizing (UMK57 and nocodazole) compounds. We discuss the utility of each technique for defining specific features of spindle microtubule dynamics and k-MT attachment stability.


Subject(s)
Cytological Techniques/methods , Kinetochores/metabolism , Microtubules/metabolism , Calcium/pharmacology , Cell Line , Cold-Shock Response/drug effects , Humans , Imaging, Three-Dimensional , Kinetochores/drug effects , Light , Microtubules/drug effects , Nocodazole/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
7.
Cell Cycle ; 18(15): 1702-1713, 2019 08.
Article in English | MEDLINE | ID: mdl-31179849

ABSTRACT

Chromosomal instability (CIN) is defined as a high rate of whole chromosome loss or gain and is a hallmark of many aneuploid solid tumors. CIN positively correlates with poor patient prognosis and chemotherapeutic resistance. Despite this clinical importance, the role of CIN in tumor initiation, growth and/or progression remains poorly understood. To date, the only strategies developed to determine how CIN contributes to tumorigenesis have relied on transgenic mouse models that deliberately increase the rate of chromosomal mis-segregation. Here we develop a strain of transgenic mice that is designed to strategically decrease the rate of chromosome mis-segregation and suppress CIN. These animals modestly overexpress the kinesin-13 microtubule depolymerase Kif2b, a strategy proven successful in restoring faithful chromosome segregation to human cancer cells in culture. Using the LA2 K-Ras G12D-induced model for lung cancer, we show that Kif2b expression reduces the number of chromosome segregation defects but does not change the incidence of lung tumor lesions. However, pulmonary tumors were significantly larger in animals expressing Kif2b and those tumors exhibited elevated rates of Ki-67 positive cells relative to controls. Thus, in lung cancers driven by mutations in K-Ras, CIN has little impact on tumor initiation but suppresses tumor growth. These data support a model in which CIN imposes a burden on tumor cells, and that enhancement of mitotic fidelity results in accelerated tumor growth.


Subject(s)
Adenoma/genetics , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/genetics , Kinesins/metabolism , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Adenoma/metabolism , Adenoma/pathology , Aneuploidy , Animals , Cell Line , Cell Transformation, Neoplastic/metabolism , Chromosome Segregation/genetics , Fibroblasts , Kinesins/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/genetics , Up-Regulation
8.
BMC Med Genomics ; 12(1): 79, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31151460

ABSTRACT

BACKGROUND: Intra-tumor heterogeneity stems from genetic, epigenetic, functional, and environmental differences among tumor cells. A major source of genetic heterogeneity comes from DNA sequence differences and/or whole chromosome and focal copy number variations (CNVs). Whole chromosome CNVs are caused by chromosomal instability (CIN) that is defined by a persistently high rate of chromosome mis-segregation. Accordingly, CIN causes constantly changing karyotypes that result in extensive cell-to-cell genetic heterogeneity. How the genetic heterogeneity caused by CIN influences gene expression in individual cells remains unknown. METHODS: We performed single-cell RNA sequencing on a chromosomally unstable glioblastoma cancer stem cell (CSC) line and a control normal, diploid neural stem cell (NSC) line to investigate the impact of CNV due to CIN on gene expression. From the gene expression data, we computationally inferred large-scale CNVs in single cells. Also, we performed copy number adjusted differential gene expression analysis between NSCs and glioblastoma CSCs to identify copy number dependent and independent differentially expressed genes. RESULTS: Here, we demonstrate that gene expression across large genomic regions scales proportionally to whole chromosome copy number in chromosomally unstable CSCs. Also, we show that the differential expression of most genes between normal NSCs and glioblastoma CSCs is largely accounted for by copy number alterations. However, we identify 269 genes whose differential expression in glioblastoma CSCs relative to normal NSCs is independent of copy number. Moreover, a gene signature derived from the subset of genes that are differential expressed independent of copy number in glioblastoma CSCs correlates with tumor grade and is prognostic for patient survival. CONCLUSIONS: These results demonstrate that CIN is directly responsible for gene expression changes and contributes to both genetic and transcriptional heterogeneity among glioblastoma CSCs. These results also demonstrate that the expression of some genes is buffered against changes in copy number, thus preserving some consistency in gene expression levels from cell-to-cell despite the continuous change in karyotype driven by CIN. Importantly, a gene signature derived from the subset of genes whose expression is buffered against copy number alterations correlates with tumor grade and is prognostic for patient survival that could facilitate patient diagnosis and treatment.


Subject(s)
Chromosomal Instability , Glioblastoma/genetics , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Cell Line, Tumor , Gene Expression Profiling , Humans , Neoplasm Grading , Neoplastic Stem Cells/pathology , Neural Stem Cells/metabolism , Survival Analysis
9.
Methods Cell Biol ; 144: 15-32, 2018.
Article in English | MEDLINE | ID: mdl-29804667

ABSTRACT

Cell viability requires accurate chromosome segregation during meiosis and mitosis so that the daughter cells produced have the correct chromosome complement. In contrast, chromosome segregation errors lead to aneuploidy, a state of abnormal chromosome numbers. Furthermore, a persistently high rate of chromosome segregation errors causes the related phenomenon of whole chromosomal instability (w-CIN). Aneuploidy and w-CIN are common characteristics of several human conditions and diseases including birth defects and cancers. Thus, methods to measure aneuploidy and w-CIN have important research applications in many areas of cell biology. In this chapter, we describe methods to measure chromosome missegregation rates and aneuploid cell survival with a focus on cells grown in culture; however, we also highlight methods that are amenable to primary tissue samples. Together, these methods provide a comprehensive approach to determining the frequency of aneuploidy and w-CIN in cells.


Subject(s)
Aneuploidy , Chromosomal Instability/genetics , Cytological Techniques/methods , Biological Assay , Cell Line, Tumor , Cell Survival , Chromosome Segregation , Chromosomes, Human/metabolism , Fluorescent Antibody Technique , Humans , In Situ Hybridization, Fluorescence , Mitosis
10.
Elife ; 62017 11 20.
Article in English | MEDLINE | ID: mdl-29154753

ABSTRACT

The fidelity of chromosome segregation in mitosis is safeguarded by the precise regulation of kinetochore microtubule (k-MT) attachment stability. Previously, we demonstrated that Cyclin A/Cdk1 destabilizes k-MT attachments to promote faithful chromosome segregation. Here, we use quantitative phosphoproteomics to identify 156 Cyclin A/Cdk1 substrates in prometaphase. One Cyclin A/Cdk1 substrate is myosin phosphatase targeting subunit 1 (MYPT1), and we show that MYPT1 localization to kinetochores depends on Cyclin A/Cdk1 activity and that MYPT1 destabilizes k-MT attachments by negatively regulating Plk1 at kinetochores. Thus, Cyclin A/Cdk1 phosphorylation primes MYPT1 for Plk1 binding. Interestingly, priming of PBIP1 by Plk1 itself (self-priming) increased in MYPT1-depleted cells showing that MYPT1 provides a molecular link between the processes of Cdk1-dependent priming and self-priming of Plk1 substrates. These data demonstrate cross-regulation between Cyclin A/Cdk1-dependent and Plk1-dependent phosphorylation of substrates during mitosis to ensure efficient correction of k-MT attachment errors necessary for high mitotic fidelity.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cyclin A/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Prometaphase , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Cell Line , Chromosome Segregation , Humans , Phosphorylation , Protein Processing, Post-Translational , Polo-Like Kinase 1
11.
Trends Cancer ; 3(3): 225-234, 2017 03.
Article in English | MEDLINE | ID: mdl-28718433

ABSTRACT

DNA double-strand breaks (DSBs) prevent cells from entering mitosis allowing cells to repair their genomic damage. Little is known about the response to DSBs once cells have already committed to mitosis. Here, we review the genome-protective role of the mitotic DNA damage response (DDR) and evidence suggesting that its untimely activation induces chromosome segregation errors and paradoxically undermines genomic integrity. In contrast to normal cells, cancer cells coopt this pathway to propagate structural and numerical chromosomal instabilities. Cells derived from genomically unstable tumors exhibit evidence for a partially activated DDR during mitosis, which leads to ongoing chromosome segregation errors. Thus, a thorough understanding of the consequences of mitotic DNA damage is key to our ability to devise novel anticancer therapeutic strategies.


Subject(s)
Chromosomal Instability/genetics , Mitosis/genetics , Neoplasms/genetics , Chromosome Segregation/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA Repair/genetics , Humans , Neoplasms/pathology
12.
Cell Rep ; 17(7): 1755-1763, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27829147

ABSTRACT

Karyotype diversity is a hallmark of solid tumors that contributes to intratumor heterogeneity. This diversity is generated by persistent chromosome mis-segregation associated with chromosomal instability (CIN). CIN correlates with tumor relapse and is thought to promote drug resistance by creating a vast genomic landscape through which karyotypically unique clones survive lethal drug selection. We explore this proposition using a small molecule (UMK57) that suppresses chromosome mis-segregation in CIN cancer cells by potentiating the activity of the kinesin-13 protein MCAK. Sublethal doses of UMK57 destabilize kinetochore-microtubule (k-MT) attachments during mitosis to increase chromosome segregation fidelity. Surprisingly, chromosome mis-segregation rebounds in UMK57-treated cancer cells within a few days. This rapid relapse is driven by alterations in the Aurora B signaling pathway that hyper-stabilize k-MT attachments and is reversible following UMK57 removal. Thus, cancer cells display adaptive resistance to therapies targeting CIN through rapid and reversible changes to mitotic signaling networks.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosomal Instability/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/pathology , Aurora Kinase B/metabolism , Cell Line, Tumor , Chromosomes, Human/metabolism , Humans , Kinesins/metabolism
13.
Mol Cancer Ther ; 15(11): 2758-2766, 2016 11.
Article in English | MEDLINE | ID: mdl-27550941

ABSTRACT

Despite advances in targeted therapy, lung cancer remains the most common cause of cancer-related mortality in the United States. Chromosomal instability is a prominent feature in lung cancer and, because it rarely occurs in normal cells, it represents a potential therapeutic target. Our prior work discovered that lung cancer cells undergo anaphase catastrophe in response to inhibition of cyclin-dependent kinase 2 (CDK2), followed by apoptosis and reduced growth. In this study, the effects and mechanisms of the multi-CDK inhibitor dinaciclib on lung cancer cells were investigated. We sought to determine the specificity of CDK-dependent induction of anaphase catastrophe. Live cell imaging provided direct evidence that dinaciclib caused multipolar cell divisions resulting in extensive chromosome missegregation. Genetic knockdown of dinaciclib CDK targets revealed that repression of CDK2 and CDK1, but not CDK5 or CDK9, triggered anaphase catastrophe in lung cancer cells. Overexpression of CP110, which is a mediator of CDK2 inhibitor-induced anaphase catastrophe (and a CDK1 and 2 phosphorylation substrate), antagonized anaphase catastrophe and apoptosis following dinaciclib treatment. Consistent with our previous findings, acquisition of activated KRAS sensitized lung cancer cells to dinaciclib-mediated anaphase catastrophe and cell death. Combining dinaciclib with the mitotic inhibitor taxol augmented anaphase catastrophe induction and reduced cell viability of lung cancer cells. Thus, the multi-CDK inhibitor dinaciclib causes anaphase catastrophe in lung cancer cells and should be investigated as a potential therapeutic for wild-type and KRAS-mutant lung cancer, individually or in combination with taxanes. Mol Cancer Ther; 15(11); 2758-66. ©2016 AACR.


Subject(s)
Anaphase/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CDC2 Protein Kinase/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , Animals , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic N-Oxides , Drug Resistance, Neoplasm/genetics , Humans , Indolizines , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Mutation , Phosphoproteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Taxoids/pharmacology
14.
Cancer Discov ; 6(5): 532-45, 2016 05.
Article in English | MEDLINE | ID: mdl-27001151

ABSTRACT

UNLABELLED: Tumors are dynamic organs that evolve during disease progression with genetic, epigenetic, and environmental differences among tumor cells serving as the foundation for selection and evolution in tumors. Tumor-initiating cells (TIC) that are responsible for tumorigenesis are a source of functional cellular heterogeneity, whereas chromosomal instability (CIN) is a source of karyotypic genetic diversity. However, the extent that CIN contributes to TIC genetic diversity and its relationship to TIC function remains unclear. Here, we demonstrate that glioblastoma TICs display CIN with lagging chromosomes at anaphase and extensive nonclonal chromosome copy-number variations. Elevating the basal chromosome missegregation rate in TICs decreases both proliferation and the stem-like phenotype of TICs in vitro Consequently, tumor formation is abolished in an orthotopic mouse model. These results demonstrate that TICs generate genetic heterogeneity within tumors, but that TIC function is impaired if the rate of genetic change is elevated above a tolerable threshold. SIGNIFICANCE: Genetic heterogeneity among TICs may produce advantageous karyotypes that lead to therapy resistance and relapse; however, we found that TICs have an upper tolerable limit for CIN. Thus, increasing the chromosome missegregation rate offers a new therapeutic strategy to eliminate TICs from tumors. Cancer Discov; 6(5); 532-45. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 461.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomal Instability , Glioblastoma/genetics , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Chromosome Aberrations , Chromosome Segregation , DNA Fragmentation , Disease Models, Animal , Female , Genetic Heterogeneity , Genetic Predisposition to Disease , Glioblastoma/metabolism , Heterografts , Humans , In Situ Hybridization, Fluorescence , Mice , Mutation
15.
PLoS Genet ; 12(2): e1005865, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26871722

ABSTRACT

Somatic mutations of the cohesin complex subunit STAG2 are present in diverse tumor types. We and others have shown that STAG2 inactivation can lead to loss of sister chromatid cohesion and alterations in chromosome copy number in experimental systems. However, studies of naturally occurring human tumors have demonstrated little, if any, correlation between STAG2 mutational status and aneuploidy, and have further shown that STAG2-deficient tumors are often euploid. In an effort to provide insight into these discrepancies, here we analyze the effect of tumor-derived STAG2 mutations on the protein composition of cohesin and the expected mitotic phenotypes of STAG2 mutation. We find that many mutant STAG2 proteins retain their ability to interact with cohesin; however, the presence of mutant STAG2 resulted in a reduction in the ability of regulatory subunits WAPL, PDS5A, and PDS5B to interact with the core cohesin ring. Using AAV-mediated gene targeting, we then introduced nine tumor-derived mutations into the endogenous allele of STAG2 in cultured human cells. While all nonsense mutations led to defects in sister chromatid cohesion and a subset induced anaphase defects, missense mutations behaved like wild-type in these assays. Furthermore, only one of nine tumor-derived mutations tested induced overt alterations in chromosome counts. These data indicate that not all tumor-derived STAG2 mutations confer defects in cohesion, chromosome segregation, and ploidy, suggesting that there are likely to be other functional effects of STAG2 inactivation in human cancer cells that are relevant to cancer pathogenesis.


Subject(s)
Anaphase , Antigens, Nuclear/genetics , Chromosome Segregation , Mutation/genetics , Neoplasms/genetics , Base Sequence , Cell Cycle Proteins/metabolism , Cell Proliferation , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/genetics , Codon, Nonsense/genetics , Dependovirus/metabolism , Gene Targeting , HCT116 Cells , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Protein Binding , Protein Subunits/metabolism , Cohesins
16.
Mol Cancer Ther ; 14(11): 2576-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26304236

ABSTRACT

Chromosomal instability (CIN) is a hallmark of solid tumor biology and is implicated in carcinogenesis. Preferentially eliminating malignant cells by targeting CIN and aneuploidy is an attractive antineoplastic strategy. We previously reported that CDK2 antagonism causes lung cancer cells to undergo anaphase catastrophe and apoptosis through inhibition of phosphorylation of the centrosomal protein CP110. Cells with activating KRAS mutations were particularly sensitive to CDK2 inhibition due to downregulation of CP110 protein levels. This study investigated mechanisms of CDK2 antagonism that mediate anaphase catastrophe via changes in CP110 protein expression and how activated KRAS affects CP110 levels in lung cancers. Site-directed mutagenesis revealed candidate CDK phosphorylation sites of CP110 (residues Ser 170 and Thr 194) critical for conferring anaphase catastrophe by altering centrosome clustering in mitosis. Intriguingly, KRAS mutation can promote CP110 protein degradation by upregulating the ubiquitin ligase SCF(cyclinF), which targets CP110 protein for destabilization. Finally, CDK2 inhibitor response was enhanced when combined with knockdown of the deubiquitinase USP33 that in turn accelerates CP110 protein degradation. Thus, this study provides molecular pharmacologic insights into how CP110 expression regulates response to CDK2 inhibition. An improved understanding of in vitro antineoplastic mechanisms of combining CDK2 antagonism with induced CP110 repression provides a rationale for exploring clinical consequences of this strategy. Taken together, preclinical findings obtained from combining CDK2 inhibition with USP33 repression have implications for treating patients with non-small cell lung cancers.


Subject(s)
Anaphase/drug effects , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Ubiquitin Thiolesterase/metabolism , Anaphase/genetics , Animals , Binding Sites/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Centrosome/drug effects , Centrosome/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclins/metabolism , Humans , Immunoblotting , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microtubule-Associated Proteins/genetics , Mutation , Phosphoproteins/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Purines/pharmacology , RNA Interference , Roscovitine , Serine/genetics , Serine/metabolism , Threonine/genetics , Threonine/metabolism , Ubiquitin Thiolesterase/genetics
17.
Cell Rep ; 11(4): 508-15, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25892238

ABSTRACT

Correction of faulty kinetochore-microtubule attachments is essential for faithful chromosome segregation and dictated by the opposing activities of Aurora B kinase and PP1 and PP2A phosphatases. How kinase and phosphatase activities are appropriately balanced is less clear. Here, we show that a centromeric pool of PP2A-B56 counteracts Aurora B T-loop phosphorylation and is recruited to centromeres through Shugoshin-1 (Sgo1). In non-transformed RPE-1 cells, Aurora B, Sgo1, and PP2A-B56 are enriched on centromeres and levels diminish as chromosomes establish bi-oriented attachments. Elevating Sgo1 levels at centromeres recruits excess PP2A-B56, and this counteracts Aurora B kinase activity, undermining efficient correction of kinetochore-microtubule attachment errors. Conversely, Sgo1-depleted cells display reduced centromeric localization of Aurora B, whereas the remaining kinase is hyperactive due to concomitant reduction of centromeric PP2A-B56. Our data suggest that Sgo1 can tune the stability of kinetochore-microtubule attachments through recruitment of PP2A-B56 that balances Aurora B activity at the centromere.


Subject(s)
Aurora Kinase B/metabolism , Cell Cycle Proteins/metabolism , Centromere/metabolism , Protein Phosphatase 2/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Chromosome Pairing , Humans
18.
Cancer Res ; 75(10): 2029-38, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25808870

ABSTRACT

Aneuploidy is frequently detected in human cancers and is implicated in carcinogenesis. Pharmacologic targeting of aneuploidy is an attractive therapeutic strategy, as this would preferentially eliminate malignant over normal cells. We previously discovered that CDK2 inhibition causes lung cancer cells with more than two centrosomes to undergo multipolar cell division leading to apoptosis, defined as anaphase catastrophe. Cells with activating KRAS mutations were especially sensitive to CDK2 inhibition. Mechanisms of CDK2-mediated anaphase catastrophe and how activated KRAS enhances this effect were investigated. Live-cell imaging provided direct evidence that following CDK2 inhibition, lung cancer cells develop multipolar anaphase and undergo multipolar cell division with the resulting progeny apoptotic. The siRNA-mediated repression of the CDK2 target and centrosome protein CP110 induced anaphase catastrophe of lung cancer cells. In contrast, CP110 overexpression antagonized CDK2 inhibitor-mediated anaphase catastrophe. Furthermore, activated KRAS mutations sensitized lung cancer cells to CDK2 inhibition by deregulating CP110 expression. Thus, CP110 is a critical mediator of CDK2 inhibition-driven anaphase catastrophe. Independent examination of murine and human paired normal-malignant lung tissues revealed marked upregulation of CP110 in malignant versus normal lung. Human lung cancers with KRAS mutations had significantly lower CP110 expression as compared with KRAS wild-type cancers. Thus, a direct link was found between CP110 and CDK2 inhibitor antineoplastic response. CP110 plays a mechanistic role in response of lung cancer cells to CDK2 inhibition, especially in the presence of activated KRAS mutations.


Subject(s)
Anaphase/drug effects , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Phosphoproteins/metabolism , Purines/pharmacology , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 2/metabolism , Humans , Lung Neoplasms/drug therapy , Mice , Microtubule-Associated Proteins/genetics , Mutation , Phosphoproteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Roscovitine , ras Proteins/genetics
19.
Nat Commun ; 6: 5990, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25606712

ABSTRACT

The exquisite sensitivity of mitotic cancer cells to ionizing radiation (IR) underlies an important rationale for the widely used fractionated radiation therapy. However, the mechanism for this cell cycle-dependent vulnerability is unknown. Here we show that treatment with IR leads to mitotic chromosome segregation errors in vivo and long-lasting aneuploidy in tumour-derived cell lines. These mitotic errors generate an abundance of micronuclei that predispose chromosomes to subsequent catastrophic pulverization thereby independently amplifying radiation-induced genome damage. Experimentally suppressing whole-chromosome missegregation reduces downstream chromosomal defects and significantly increases the viability of irradiated mitotic cells. Further, orthotopically transplanted human glioblastoma tumours in which chromosome missegregation rates have been reduced are rendered markedly more resistant to IR, exhibiting diminished markers of cell death in response to treatment. This work identifies a novel mitotic pathway for radiation-induced genome damage, which occurs outside of the primary nucleus and augments chromosomal breaks. This relationship between radiation treatment and whole-chromosome missegregation can be exploited to modulate therapeutic response in a clinically relevant manner.


Subject(s)
Brain Neoplasms/genetics , Chromosomal Instability , Glioblastoma/genetics , Neoplasms/radiotherapy , Aneuploidy , Animals , Brain Neoplasms/radiotherapy , Cell Cycle , Cell Death , Cell Line, Tumor , Cell Survival , Chromosome Breakage , Chromosome Segregation , Glioblastoma/radiotherapy , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Micronucleus Tests , Mitosis/genetics , Neoplasm Transplantation , Radiation, Ionizing
20.
Nat Rev Mol Cell Biol ; 16(1): 57-64, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25466864

ABSTRACT

Faithful chromosome segregation during mitosis is essential for genome integrity and is mediated by the bi-oriented attachment of replicated chromosomes to spindle microtubules through kinetochores. Errors in kinetochore-microtubule (k-MT) attachment that could cause chromosome mis-segregation are frequent and are corrected by the dynamic turnover of k-MT attachments. Thus, regulating the rate of spindle microtubule attachment and detachment to kinetochores is crucial for mitotic fidelity and is frequently disrupted in cancer cells displaying chromosomal instability. A model based on homeostatic principles involving receptors, a core control network, effectors and feedback control may explain the precise regulation of k-MT attachment stability during mitotic progression to ensure error-free mitosis.


Subject(s)
Chromosomal Instability , Chromosome Segregation , Kinetochores/metabolism , Microtubules/metabolism , Mitosis , Neoplasms/metabolism , Animals , Humans , Microtubules/genetics , Neoplasms/genetics , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...