Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cell ; 154(2): 269-71, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870117

ABSTRACT

Development and homeostasis require repeated symmetric cell divisions, which in turn depend on a centered mitotic spindle. In this issue, Kiyomitsu and Cheeseman uncover two mechanisms that ensure correct spindle positioning in anaphase: cortical dynein for pulling the spindle to the cell center and asymmetric membrane elongation that adjusts the position of the cell center to the cleavage plane.


Subject(s)
Anaphase , Cell Membrane/metabolism , Dyneins/metabolism , Spindle Apparatus/metabolism , Animals , Humans
2.
Nat Chem Biol ; 15(8): 822-829, 2019 08.
Article in English | MEDLINE | ID: mdl-31285596

ABSTRACT

Here, we report the fragment-based discovery of BI-9321, a potent, selective and cellular active antagonist of the NSD3-PWWP1 domain. The human NSD3 protein is encoded by the WHSC1L1 gene located in the 8p11-p12 amplicon, frequently amplified in breast and squamous lung cancer. Recently, it was demonstrated that the PWWP1 domain of NSD3 is required for the viability of acute myeloid leukemia cells. To further elucidate the relevance of NSD3 in cancer biology, we developed a chemical probe, BI-9321, targeting the methyl-lysine binding site of the PWWP1 domain with sub-micromolar in vitro activity and cellular target engagement at 1 µM. As a single agent, BI-9321 downregulates Myc messenger RNA expression and reduces proliferation in MOLM-13 cells. This first-in-class chemical probe BI-9321, together with the negative control BI-9466, will greatly facilitate the elucidation of the underexplored biological function of PWWP domains.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , CRISPR-Cas Systems , Cell Line , Cell Proliferation/drug effects , Cell Survival , Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
3.
Future Oncol ; 15(6): 637-652, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30404555

ABSTRACT

While the development of EGFR-targeted tyrosine kinase inhibitors (TKIs) has revolutionized treatment of EGFR mutation-positive non-small-cell lung cancer, acquired resistance to therapy is inevitable, reflecting tumor evolution. Recent studies show that EGFR mutation-positive non-small-cell lung cancer is highly heterogeneous at the cellular level, facilitating clonal expansion of resistant tumors via multiple molecular mechanisms. Here, we review the mechanistic differences between first-, second- and third-generation EGFR-targeted TKIs and speculate how these features could explain differences in clinical activity between these agents from a clonal evolution perspective. We hypothesize that the molecular dissection of tumor resistance mechanisms will facilitate optimal sequential use of EGFR TKIs in individual patients, thus maximizing the duration of chemotherapy-free treatment and survival benefit.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Clonal Evolution/genetics , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Mutation , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/therapy , ErbB Receptors/genetics , Genetic Heterogeneity , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/therapy , Molecular Targeted Therapy
4.
EMBO J ; 33(22): 2623-42, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25257310

ABSTRACT

Sister chromatid cohesion mediated by the cohesin complex is essential for chromosome segregation during cell division. Using functional genomic screening, we identify a set of 26 pre-mRNA splicing factors that are required for sister chromatid cohesion in human cells. Loss of spliceosome subunits increases the dissociation rate of cohesin from chromatin and abrogates cohesion after DNA replication, ultimately causing mitotic catastrophe. Depletion of splicing factors causes defective processing of the pre-mRNA encoding sororin, a factor required for the stable association of cohesin with chromatin, and an associated reduction of sororin protein level. Expression of an intronless version of sororin and depletion of the cohesin release protein WAPL suppress the cohesion defect in cells lacking splicing factors. We propose that spliceosome components contribute to sister chromatid cohesion and mitotic chromosome segregation through splicing of sororin pre-mRNA. Our results highlight the loss of cohesion as an early cellular consequence of compromised splicing. This may have clinical implications because SF3B1, a splicing factor that we identify to be essential for cohesion, is recurrently mutated in chronic lymphocytic leukaemia.


Subject(s)
Chromatids , Leukemia, Lymphocytic, Chronic, B-Cell , Mutation , Neoplasm Proteins , Phosphoproteins , RNA Splicing , RNA, Neoplasm , Ribonucleoprotein, U2 Small Nuclear , Sister Chromatid Exchange , Chromatids/genetics , Chromatids/metabolism , Genomics/methods , HeLa Cells , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing Factors , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism
5.
Nature ; 492(7428): 276-9, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23235882

ABSTRACT

At the end of cell division, cytokinesis splits the cytoplasm of nascent daughter cells and partitions segregated sister genomes. To coordinate cell division with chromosome segregation, the mitotic spindle controls cytokinetic events at the cell envelope. The spindle midzone stimulates the actomyosin-driven contraction of the cleavage furrow, which proceeds until the formation of a microtubule-rich intercellular bridge with the midbody at its centre. The midbody directs the final membrane abscission reaction and has been proposed to attach the cleavage furrow to the intercellular bridge. How the mitotic spindle is connected to the plasma membrane during cytokinesis is not understood. Here we identify a plasma membrane tethering activity in the centralspindlin protein complex, a conserved component of the spindle midzone and midbody. We demonstrate that the C1 domain of the centralspindlin subunit MgcRacGAP associates with the plasma membrane by interacting with polyanionic phosphoinositide lipids. Using X-ray crystallography we determine the structure of this atypical C1 domain. Mutations in the hydrophobic cap and in basic residues of the C1 domain of MgcRacGAP prevent association of the protein with the plasma membrane, and abrogate cytokinesis in human and chicken cells. Artificial membrane tethering of centralspindlin restores cell division in the absence of the C1 domain of MgcRacGAP. Although C1 domain function is dispensable for the formation of the midzone and midbody, it promotes contractility and is required for the attachment of the plasma membrane to the midbody, a long-postulated function of this organelle. Our analysis suggests that centralspindlin links the mitotic spindle to the plasma membrane to secure the final cut during cytokinesis in animal cells.


Subject(s)
Cell Membrane/metabolism , Cytokinesis/radiation effects , GTPase-Activating Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Cytokinesis/genetics , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubules/chemistry , Microtubules/metabolism , Models, Molecular , Protein Binding , Protein Kinase C-alpha/metabolism , Protein Structure, Tertiary , Protein Transport/drug effects , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology
6.
Clin Cancer Res ; 30(8): 1582-1594, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38330145

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) serve as the standard first-line therapy for EGFR-mutated non-small cell lung cancer (NSCLC). Despite the sustained clinical benefits achieved through optimal EGFR-TKI treatments, including the third-generation EGFR-TKI osimertinib, resistance inevitably develops. Currently, there are no targeted therapeutic options available postprogression on osimertinib. Here, we assessed the preclinical efficacy of BI-4732, a novel fourth-generation EGFR-TKI, using patient-derived preclinical models reflecting various clinical scenarios. EXPERIMENTAL DESIGN: The antitumor activity of BI-4732 was evaluated using Ba/F3 cells and patient-derived cell/organoid/xenograft models with diverse EGFR mutations. Intracranial antitumor activity of BI-4732 was evaluated in a brain-metastasis mouse model. RESULTS: We demonstrated the remarkable antitumor efficacy of BI-4732 as a single agent in various patient-derived models with EGFR_C797S-mediated osimertinib resistance. Moreover, BI-4732 exhibited activity comparable to osimertinib in inhibiting EGFR-activating (E19del and L858R) and T790M mutations. In a combination treatment strategy with osimertinib, BI-4732 exhibited a synergistic effect at significantly lower concentrations than those used in monotherapy. Importantly, BI-4732 displayed potent antitumor activity in an intracranial model, with low efflux at the blood-brain barrier. CONCLUSIONS: Our findings highlight the potential of BI-4732, a selective EGFR-TKI with high blood-brain barrier penetration, targeting a broad range of EGFR mutations, including C797S, warranting clinical development.


Subject(s)
Acrylamides , Carcinoma, Non-Small-Cell Lung , Indoles , Lung Neoplasms , Pyrimidines , Mice , Animals , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , ErbB Receptors/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mutation , Drug Resistance, Neoplasm/genetics , Aniline Compounds
7.
bioRxiv ; 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36747713

ABSTRACT

Efforts to improve the anti-tumor response to KRASG12C targeted therapy have benefited from leveraging combination approaches. Here, we compare the anti-tumor response induced by the SOS1-KRAS interaction inhibitor, BI-3406, combined with a KRASG12C inhibitor (KRASG12Ci) to those induced by KRASG12Ci alone or combined with SHP2 or EGFR inhibitors. In lung cancer and colorectal cancer (CRC) models, BI-3406 plus KRASG12Ci induces an anti-tumor response stronger than that observed with KRASG12Ci alone and comparable to those by the other combinations. This enhanced anti-tumor response is associated with a stronger and extended suppression of RAS-MAPK signaling. Importantly, BI-3406 plus KRASG12Ci treatment delays the emergence of acquired adagrasib resistance in both CRC and lung cancer models and is associated with re-establishment of anti-proliferative activity in KRASG12Ci-resistant CRC models. Our findings position KRASG12C plus SOS1 inhibition therapy as a promising strategy for treating both KRASG12C-mutated tumors as well as for addressing acquired resistance to KRASG12Ci.

8.
Dev Cell ; 12(5): 713-25, 2007 May.
Article in English | MEDLINE | ID: mdl-17488623

ABSTRACT

Cytokinesis of animal cells requires ingression of the actomyosin-based contractile ring between segregated sister genomes. Localization of the RhoGEF Ect2 to the central spindle at anaphase promotes local activation of the RhoA GTPase, which induces assembly and ingression of the contractile ring. Here we have used BI 2536, an inhibitor of the mitotic kinase Plk1, to analyze the functions of this enzyme during late mitosis in human cells. We show that Plk1 acts after Cdk1 inactivation and independently from Aurora B to promote RhoA accumulation at the equator, contractile ring formation, and cleavage furrow ingression. Inhibition of Plk1 abolishes the interaction of Ect2 with its activator and midzone anchor, HsCyk-4, thereby preventing localization of Ect2 to the central spindle. We propose that late mitotic Plk1 activity promotes recruitment of Ect2 to the central spindle, triggering the initiation of cytokinesis and contributing to cleavage plane specification in human cells.


Subject(s)
Cell Cycle Proteins/metabolism , Cytokinesis , Guanine Nucleotide Exchange Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Spindle Apparatus/metabolism , Aurora Kinase B , Aurora Kinases , CDC2 Protein Kinase/metabolism , Catalysis/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Chromosomes, Human/genetics , Cytokinesis/drug effects , Enzyme Activation/drug effects , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Models, Biological , Protein Binding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Transport/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Rho Guanine Nucleotide Exchange Factors , Spindle Apparatus/drug effects , rhoA GTP-Binding Protein/metabolism , Polo-Like Kinase 1
9.
J Cell Sci ; 123(Pt 9): 1395-400, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20356927

ABSTRACT

Germline mutations in the tumor-suppressor gene BRCA2 predispose to breast and ovarian cancer. BRCA2 plays a well-established role in maintaining genome stability by regulating homologous recombination. BRCA2 has more recently been implicated in cytokinesis, the final step of cell division, but the molecular basis for this remains unknown. We have used time-lapse microscopy, recently developed cytokinesis assays and BAC recombineering (bacterial artificial chromosome recombinogenic engineering) to investigate the function and localization of BRCA2 during cell division. Our analysis suggests that BRCA2 does not regulate cytokinesis in human cells. Thus, cytokinesis defects are unlikely to contribute to chromosomal instability and tumorigenesis in BRCA2-related cancers.


Subject(s)
BRCA2 Protein/metabolism , Cytokinesis , Apoptosis Regulatory Proteins , Cell Nucleus/metabolism , Chromosomes, Artificial, Bacterial/metabolism , Gene Targeting , HeLa Cells , Humans , Microtubules/metabolism , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism , Spindle Apparatus/metabolism , Time Factors , Transfection
10.
PLoS Biol ; 7(5): e1000110, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19468300

ABSTRACT

To complete cell division with high fidelity, cytokinesis must be coordinated with chromosome segregation. Mammalian Polo-like kinase 1, Plk1, may function as a critical link because it is required for chromosome segregation and establishment of the cleavage plane following anaphase onset. A central spindle-localized pool of the RhoGEF Ect2 promotes activation of the small GTPase RhoA, which drives contractile ring assembly at the equatorial cortex. Here, we have investigated how Plk1 promotes the central spindle recruitment of Ect2. Plk1 phosphorylates the noncatalytic N terminus of the RhoGAP HsCyk-4 at the central spindle, creating a phospho-epitope recognized by the BRCA1 C-terminal (BRCT) repeats of Ect2. Failure to phosphorylate HsCyk-4 blocks Ect2 recruitment to the central spindle and the subsequent induction of furrowing. Microtubules, as well as the microtubule-associated protein (MAP) Prc1, facilitate Plk1 phosphorylation of HsCyk-4. Characterization of a phosphomimetic version of HsCyk-4 indicates that Plk1 promotes Ect2 recruitment through multiple targets. Collectively, our data reveal that formation of the HsCyk-4-Ect2 complex is subject to multiple layers of regulation to ensure that RhoA activation occurs between the segregated sister chromatids during anaphase.


Subject(s)
Cell Cycle Proteins/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Biological Transport/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoblotting , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors , Serine/metabolism , Two-Hybrid System Techniques , Polo-Like Kinase 1
11.
Nature ; 441(7089): 53-61, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16541024

ABSTRACT

Segregation of homologous maternal and paternal centromeres to opposite poles during meiosis I depends on post-replicative crossing over between homologous non-sister chromatids, which creates chiasmata and therefore bivalent chromosomes. Destruction of sister chromatid cohesion along chromosome arms due to proteolytic cleavage of cohesin's Rec8 subunit by separase resolves chiasmata and thereby triggers the first meiotic division. This produces univalent chromosomes, the chromatids of which are held together by centromeric cohesin that has been protected from separase by shugoshin (Sgo1/MEI-S332) proteins. Here we show in both fission and budding yeast that Sgo1 recruits to centromeres a specific form of protein phosphatase 2A (PP2A). Its inactivation causes loss of centromeric cohesin at anaphase I and random segregation of sister centromeres at the second meiotic division. Artificial recruitment of PP2A to chromosome arms prevents Rec8 phosphorylation and hinders resolution of chiasmata. Our data are consistent with the notion that efficient cleavage of Rec8 requires phosphorylation of cohesin and that this is blocked by PP2A at meiosis I centromeres.


Subject(s)
Centromere/metabolism , Chromatids/metabolism , Chromosome Pairing , Meiosis , Phosphoprotein Phosphatases/metabolism , Saccharomyces cerevisiae/cytology , Schizosaccharomyces/cytology , Animals , Cell Cycle Proteins/metabolism , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , Conserved Sequence , Humans , Mice , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Phosphatase 2 , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Cohesins
12.
Cancer Discov ; 12(4): 924-937, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35046095

ABSTRACT

KRAS is the most frequently mutated oncogene, harboring mutations in approximately one in seven cancers. Allele-specific KRASG12C inhibitors are currently changing the treatment paradigm for patients with KRASG12C-mutated non-small cell lung cancer and colorectal cancer. The success of addressing a previously elusive KRAS allele has fueled drug discovery efforts for all KRAS mutants. Pan-KRAS drugs have the potential to address broad patient populations, including KRASG12D-, KRASG12V-, KRASG13D-, KRASG12R-, and KRASG12A-mutant or KRAS wild-type-amplified cancers, as well as cancers with acquired resistance to KRASG12C inhibitors. Here, we review actively pursued allele-specific and pan-KRAS inhibition strategies and their potential utility. SIGNIFICANCE: Mutant-selective KRASG12C inhibitors target a fraction (approximately 13.6%) of all KRAS-driven cancers. A broad arsenal of KRAS drugs is needed to comprehensively conquer KRAS-driven cancers. Conceptually, we foresee two future classes of KRAS medicines: mutant-selective KRAS drugs targeting individual variant alleles and pan-KRAS therapeutics targeting a broad range of KRAS alterations.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Mutation , Oncogenes , Precision Medicine , Proto-Oncogene Proteins p21(ras)/genetics
13.
Nat Cancer ; 3(7): 821-836, 2022 07.
Article in English | MEDLINE | ID: mdl-35883003

ABSTRACT

Oncogenic alterations in human epidermal growth factor receptor 2 (HER2) occur in approximately 2% of patients with non-small cell lung cancer and predominantly affect the tyrosine kinase domain and cluster in exon 20 of the ERBB2 gene. Most clinical-grade tyrosine kinase inhibitors are limited by either insufficient selectivity against wild-type (WT) epidermal growth factor receptor (EGFR), which is a major cause of dose-limiting toxicity or by potency against HER2 exon 20 mutant variants. Here we report the discovery of covalent tyrosine kinase inhibitors that potently inhibit HER2 exon 20 mutants while sparing WT EGFR, which reduce tumor cell survival and proliferation in vitro and result in regressions in preclinical xenograft models of HER2 exon 20 mutant non-small cell lung cancer, concomitant with inhibition of downstream HER2 signaling. Our results suggest that HER2 exon 20 insertion-driven tumors can be effectively treated by a potent and highly selective HER2 inhibitor while sparing WT EGFR, paving the way for clinical translation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Exons/genetics , Genes, erbB-2 , Humans , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/genetics
14.
Life Sci Alliance ; 4(1)2021 01.
Article in English | MEDLINE | ID: mdl-33199508

ABSTRACT

Loss of WRN, a DNA repair helicase, was identified as a strong vulnerability of microsatellite instable (MSI) cancers, making WRN a promising drug target. We show that ATP binding and hydrolysis are required for genome integrity and viability of MSI cancer cells. We report a 2.2-Å crystal structure of the WRN helicase core (517-1,093), comprising the two helicase subdomains and winged helix domain but not the HRDC domain or nuclease domains. The structure highlights unusual features. First, an atypical mode of nucleotide binding that results in unusual relative positioning of the two helicase subdomains. Second, an additional ß-hairpin in the second helicase subdomain and an unusual helical hairpin in the Zn2+ binding domain. Modelling of the WRN helicase in complex with DNA suggests roles for these features in the binding of alternative DNA structures. NMR analysis shows a weak interaction between the HRDC domain and the helicase core, indicating a possible biological role for this association. Together, this study will facilitate the structure-based development of inhibitors against WRN helicase.


Subject(s)
Catalytic Domain , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Microsatellite Instability , Werner Syndrome Helicase/chemistry , Werner Syndrome Helicase/genetics , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Cell Cycle Proteins/genetics , Cell Survival/genetics , Crystallization , DNA/metabolism , DNA Damage/genetics , Gene Silencing , HCT116 Cells , Humans , Hydrolysis , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Transfection , Zinc/metabolism , Polo-Like Kinase 1
15.
Nat Genet ; 53(3): 269-278, 2021 03.
Article in English | MEDLINE | ID: mdl-33558760

ABSTRACT

Cancer-associated, loss-of-function mutations in genes encoding subunits of the BRG1/BRM-associated factor (BAF) chromatin-remodeling complexes1-8 often cause drastic chromatin accessibility changes, especially in important regulatory regions9-19. However, it remains unknown how these changes are established over time (for example, immediate consequences or long-term adaptations), and whether they are causative for intracomplex synthetic lethalities, abrogating the formation or activity of BAF complexes9,20-24. In the present study, we use the dTAG system to induce acute degradation of BAF subunits and show that chromatin alterations are established faster than the duration of one cell cycle. Using a pharmacological inhibitor and a chemical degrader of the BAF complex ATPase subunits25,26, we show that maintaining genome accessibility requires constant ATP-dependent remodeling. Completely abolishing BAF complex function by acute degradation of a synthetic lethal subunit in a paralog-deficient background results in an almost complete loss of chromatin accessibility at BAF-controlled sites, especially also at superenhancers, providing a mechanism for intracomplex synthetic lethalities.


Subject(s)
Chromatin/genetics , DNA Helicases/metabolism , Multiprotein Complexes/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Cell Line , Chromatin/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/cytology , Enhancer Elements, Genetic , Gene Knockout Techniques , Histones/genetics , Histones/metabolism , Humans , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics
16.
Cancer Discov ; 11(1): 142-157, 2021 01.
Article in English | MEDLINE | ID: mdl-32816843

ABSTRACT

KRAS is the most frequently mutated driver of pancreatic, colorectal, and non-small cell lung cancers. Direct KRAS blockade has proved challenging, and inhibition of a key downstream effector pathway, the RAF-MEK-ERK cascade, has shown limited success because of activation of feedback networks that keep the pathway in check. We hypothesized that inhibiting SOS1, a KRAS activator and important feedback node, represents an effective approach to treat KRAS-driven cancers. We report the discovery of a highly potent, selective, and orally bioavailable small-molecule SOS1 inhibitor, BI-3406, that binds to the catalytic domain of SOS1, thereby preventing the interaction with KRAS. BI-3406 reduces formation of GTP-loaded RAS and limits cellular proliferation of a broad range of KRAS-driven cancers. Importantly, BI-3406 attenuates feedback reactivation induced by MEK inhibitors and thereby enhances sensitivity of KRAS-dependent cancers to MEK inhibition. Combined SOS1 and MEK inhibition represents a novel and effective therapeutic concept to address KRAS-driven tumors. SIGNIFICANCE: To date, there are no effective targeted pan-KRAS therapies. In-depth characterization of BI-3406 activity and identification of MEK inhibitors as effective combination partners provide an attractive therapeutic concept for the majority of KRAS-mutant cancers, including those fueled by the most prevalent mutant KRAS oncoproteins, G12D, G12V, G12C, and G13D.See related commentary by Zhao et al., p. 17.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Lung Neoplasms , Proto-Oncogene Proteins p21(ras) , Cell Line, Tumor , Humans , Mitogen-Activated Protein Kinase Kinases , Mutation , Nucleotides , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics
17.
Curr Biol ; 17(4): 316-22, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17291758

ABSTRACT

Fine-mapping of the cell-division cycle, notably the identification of mitotic kinase signaling pathways, provides novel opportunities for cancer-drug discovery. As a key regulator of multiple steps during mitotic progression across eukaryotic species, the serine/threonine-specific Polo-like kinase 1 (Plk1) is highly expressed in malignant cells and serves as a negative prognostic marker in specific human cancer types . Here, we report the discovery of a potent small-molecule inhibitor of mammalian Plk1, BI 2536, which inhibits Plk1 enzyme activity at low nanomolar concentrations. The compound potently causes a mitotic arrest and induces apoptosis in human cancer cell lines of diverse tissue origin and oncogenome signature. BI 2536 inhibits growth of human tumor xenografts in nude mice and induces regression of large tumors with well-tolerated intravenous dose regimens. In treated tumors, cells arrest in prometaphase, accumulate phosphohistone H3, and contain aberrant mitotic spindles. This mitotic arrest is followed by a surge in apoptosis, detectable by immunohistochemistry and noninvasive optical and magnetic resonance imaging. For addressing the therapeutic potential of Plk1 inhibition, BI 2536 has progressed into clinical studies in patients with locally advanced or metastatic cancers.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle/physiology , Enzyme Inhibitors/pharmacology , Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Signal Transduction/physiology , Animals , Body Weight , Cell Cycle Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Female , Flow Cytometry , HeLa Cells , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Microscopy, Fluorescence , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pteridines/metabolism , Spectrometry, Fluorescence , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
18.
Curr Biol ; 17(4): 304-15, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17291761

ABSTRACT

BACKGROUND: The mitotic kinases, Cdk1, Aurora A/B, and Polo-like kinase 1 (Plk1) have been characterized extensively to further understanding of mitotic mechanisms and as potential targets for cancer therapy. Cdk1 and Aurora kinase studies have been facilitated by small-molecule inhibitors, but few if any potent Plk1 inhibitors have been identified. RESULTS: We describe the cellular effects of a novel compound, BI 2536, a potent and selective inhibitor of Plk1. The fact that BI 2536 blocks Plk1 activity fully and instantaneously enabled us to study controversial and unknown functions of Plk1. Cells treated with BI 2536 are delayed in prophase but eventually import Cdk1-cyclin B into the nucleus, enter prometaphase, and degrade cyclin A, although BI 2536 prevents degradation of the APC/C inhibitor Emi1. BI 2536-treated cells lack prophase microtubule asters and thus polymerize mitotic microtubules only after nuclear-envelope breakdown and form monopolar spindles that do not stably attach to kinetochores. Mad2 accumulates at kinetochores, and cells arrest with an activated spindle-assembly checkpoint. BI 2536 prevents Plk1's enrichment at kinetochores and centrosomes, and when added to metaphase cells, it induces detachment of microtubules from kinetochores and leads to spindle collapse. CONCLUSIONS: Our results suggest that Plk1's accumulation at centrosomes and kinetochores depends on its own activity and that this activity is required for maintaining centrosome and kinetochore function. Our data also show that Plk1 is not required for prophase entry, but delays transition to prometaphase, and that Emi1 destruction in prometaphase is not essential for APC/C-mediated cyclin A degradation.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Enzyme Inhibitors/pharmacology , F-Box Proteins/metabolism , Microtubules/drug effects , Mitosis/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Spindle Apparatus/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Image Processing, Computer-Assisted , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pteridines/metabolism , Spindle Apparatus/metabolism , Polo-Like Kinase 1
19.
Biochem Soc Trans ; 38(6): 1660-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118144

ABSTRACT

Two closely connected mechanisms safeguard the fidelity of chromosome segregation in eukaryotic cells. The mitotic checkpoint monitors the attachment of kinetochores to microtubules and delays anaphase onset until all sister kinetochores have become attached to opposite poles. In addition, an error correction mechanism destabilizes erroneous attachments that do not lead to tension at sister kinetochores. Aurora B kinase, the catalytic subunit of the CPC (chromosomal passenger complex), acts as a sensor and effector in both pathways. In this review we focus on a poorly understood but important aspect of mitotic control: what prevents the mitotic checkpoint from springing into action when sister centromeres are split and tension is suddenly lost at anaphase onset? Recent work has shown that disjunction of sister chromatids, in principle, engages the mitotic checkpoint, and probably also the error correction mechanism, with potentially catastrophic consequences for cell division. Eukaryotic cells have solved this 'anaphase problem' by disabling the mitotic checkpoint at the metaphase-to-anaphase transition. Checkpoint inactivation is in part due to the reversal of Cdk1 (cyclin-dependent kinase 1) phosphorylation of the CPC component INCENP (inner centromere protein; Sli15 in budding yeast), which causes the relocation of the CPC from centromeres to the spindle midzone. These findings highlight principles of mitotic checkpoint control: when bipolar chromosome attachment is reached in mitosis, the checkpoint is satisfied, but still active and responsive to loss of tension. Mitotic checkpoint inactivation at anaphase onset is required to prevent checkpoint re-engagement when sister chromatids split.


Subject(s)
Anaphase/genetics , Chromatids/metabolism , Chromosome Segregation , Spindle Apparatus/metabolism , Animals , Aurora Kinase B , Aurora Kinases , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Genes, cdc , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Stress, Mechanical , Yeasts/genetics , Yeasts/physiology
20.
Curr Biol ; 30(16): R947-R949, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32810457

ABSTRACT

Determining the site of cell cleavage is crucial for cytokinesis and involves precise activation of the RhoGEF ECT2. A new study demonstrates how a non-canonical interaction of ECT2 with centralspindlin underlies cytokinesis in animal cells, solving a mechanistic conundrum.


Subject(s)
Cytokinesis , Phosphopeptides , Animals , Cell Division , HeLa Cells , Humans , Proto-Oncogene Proteins/metabolism , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL