Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 14(9): 1179-1187, 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37736184

ABSTRACT

Cyclin-dependent kinases (CDKs) are key regulators of the cell cycle and are frequently altered in cancer cells, thereby leading to uncontrolled proliferation. In this context, CDK2 has emerged as an appealing target for anticancer drug development. Herein, we describe the discovery of a series of selective small molecule inhibitors of CDK2 beginning with historical compounds from our ERK2 program (e.g., compound 6). Structure-based drug design led to the potent and selective tool compound 32, where excellent selectivity against ERK2 and CDK4 was achieved by filling the lipophilic DFG-1 pocket and targeting interactions with CDK2-specific lower hinge binding residues, respectively. Compound 32 demonstrated 112% tumor growth inhibition in mice bearing OVCAR3 tumors with 50 mg/kg bis in die (BID) oral dosing.

2.
bioRxiv ; 2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37162991

ABSTRACT

5-fluorouracil (5-FU) is a successful and broadly used anti-cancer therapeutic. A major mechanism of action of 5-FU is thought to be through thymidylate synthase (TYMS) inhibition resulting in dTTP depletion and activation of the DNA damage response. This suggests that 5-FU should synergize with other DNA damaging agents. However, we found that combinations of 5-FU and oxaliplatin or irinotecan failed to display any evidence of synergy in clinical trials, and resulted in sub-additive killing in a panel of colorectal cancer (CRC) cell lines. In seeking to understand this antagonism, we unexpectedly found that an RNA damage response during ribosome biogenesis dominates the drug's efficacy in tumor types for which 5-FU shows clinical benefit. 5-FU has an inherent bias for RNA incorporation, and blocking this greatly reduced drug-induced lethality, indicating that accumulation of damaged RNA is more deleterious than the lack of new RNA synthesis. Using 5-FU metabolites that specifically incorporate into either RNA or DNA revealed that CRC cell lines and patient-derived colorectal cancer organoids are inherently more sensitive to RNA damage. This difference held true in cell lines from other tissues in which 5-FU has shown clinical utility, whereas cell lines from tumor tissues that lack clinical 5-FU responsiveness typically showed greater sensitivity to the drug's DNA damage effects. Analysis of changes in the phosphoproteome and ubiquitinome shows RNA damage triggers the selective ubiquitination of multiple ribosomal proteins leading to autophagy-dependent rRNA catabolism and proteasome-dependent degradation of ubiquitinated ribosome proteins. Further, RNA damage response to 5-FU is selectively enhanced by compounds that promote ribosome biogenesis, such as KDM2A inhibitors. These results demonstrate the presence of a strong RNA damage response linked to apoptotic cell death, with clear utility of combinatorially targeting this response in cancer therapy.

3.
Bioorg Med Chem Lett ; 29(16): 2294-2301, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31307887

ABSTRACT

CDK4 and CDK6 are kinases with similar sequences that regulate cell cycle progression and are validated targets in the treatment of cancer. Glioblastoma is characterized by a high frequency of CDKN2A/CCND2/CDK4/CDK6 pathway dysregulation, making dual inhibition of CDK4 and CDK6 an attractive therapeutic approach for this disease. Abemaciclib, ribociclib, and palbociclib are approved CDK4/6 inhibitors for the treatment of HR+/HER2- breast cancer, but these drugs are not expected to show strong activity in brain tumors due to poor blood brain barrier penetration. Herein, we report the identification of a brain-penetrant CDK4/6 inhibitor derived from a literature molecule with low molecular weight and topological polar surface area (MW = 285 and TPSA = 66 Å2), but lacking the CDK2/1 selectivity profile due to the absence of a basic amine. Removal of a hydrogen bond donor via cyclization of the pyrazole allowed for the introduction of basic and semi-basic amines, while maintaining in many cases efflux ratios reasonable for a CNS program. Ultimately, a basic spiroazetidine (cpKa = 8.8) was identified that afforded acceptable selectivity over anti-target CDK1 while maintaining brain-penetration in vivo (mouse Kp,uu = 0.20-0.59). To probe the potency and selectivity, our lead compound was evaluated in a panel of glioblastoma cell lines. Potency comparable to abemaciclib was observed in Rb-wild type lines U87MG, DBTRG-05MG, A172, and T98G, while Rb-deficient cell lines SF539 and M059J exhibited a lack of sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Design , Glioblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , MCF-7 Cells , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
4.
Proc Natl Acad Sci U S A ; 115(18): E4236-E4244, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29666270

ABSTRACT

Chronic inflammation is a major risk factor for colorectal cancer. The p38/MAPKAP Kinase 2 (MK2) kinase axis controls the synthesis of proinflammatory cytokines that mediate both chronic inflammation and tumor progression. Blockade of this pathway has been previously reported to suppress inflammation and to prevent colorectal tumorigenesis in a mouse model of inflammation-driven colorectal cancer, by mechanisms that are still unclear. Here, using whole-animal and tissue-specific MK2 KO mice, we show that MK2 activity in the myeloid compartment promotes tumor progression by supporting tumor neoangiogenesis in vivo. Mechanistically, we demonstrate that MK2 promotes polarization of tumor-associated macrophages into protumorigenic, proangiogenic M2-like macrophages. We further confirmed our results in human cell lines, where MK2 chemical inhibition in macrophages impairs M2 polarization and M2 macrophage-induced angiogenesis. Together, this study provides a molecular and cellular mechanism for the protumorigenic function of MK2.


Subject(s)
Colorectal Neoplasms/blood supply , Colorectal Neoplasms/epidemiology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/enzymology , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Protein Serine-Threonine Kinases/genetics
5.
Cancer Cell ; 28(5): 623-637, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26602816

ABSTRACT

In normal cells, p53 is activated by DNA damage checkpoint kinases to simultaneously control the G1/S and G2/M cell cycle checkpoints through transcriptional induction of p21(cip1) and Gadd45α. In p53-mutant tumors, cell cycle checkpoints are rewired, leading to dependency on the p38/MK2 pathway to survive DNA-damaging chemotherapy. Here we show that the RNA binding protein hnRNPA0 is the "successor" to p53 for checkpoint control. Like p53, hnRNPA0 is activated by a checkpoint kinase (MK2) and simultaneously controls both cell cycle checkpoints through distinct target mRNAs, but unlike p53, this is through the post-transcriptional stabilization of p27(Kip1) and Gadd45α mRNAs. This pathway drives cisplatin resistance in lung cancer, demonstrating the importance of post-transcriptional RNA control to chemotherapy response.


Subject(s)
Cell Cycle Checkpoints/genetics , Drug Resistance, Neoplasm/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mutation , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Aged , Animals , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cisplatin/therapeutic use , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Gene Expression Regulation, Neoplastic , Genetic Pleiotropy , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Male , Mice, Inbred C57BL , Middle Aged , Neoplasms/drug therapy , Neoplasms/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
7.
Mol Cell ; 50(2): 250-60, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23622515

ABSTRACT

Eukaryotic cell division is controlled by cyclin-dependent kinases (CDKs), which require phosphorylation by a CDK-activating kinase (CAK) for full activity. Chemical genetics uncovered requirements for the metazoan CAK Cdk7 in determining cyclin specificity and activation order of Cdk2 and Cdk1 during S and G2 phases. It was unknown if Cdk7 also activates Cdk4 and Cdk6 to promote passage of the restriction (R) point, when continued cell-cycle progression becomes mitogen independent, or if CDK-activating phosphorylation regulates G1 progression. Here we show that Cdk7 is a Cdk4- and Cdk6-activating kinase in human cells, required to maintain activity, not just to establish the active state, as is the case for Cdk1 and Cdk2. Activating phosphorylation of Cdk7 rises concurrently with that of Cdk4 as cells exit quiescence and accelerates Cdk4 activation in vitro. Therefore, mitogen signaling drives a CDK-activation cascade during G1 progression, and CAK might be rate-limiting for R point passage.


Subject(s)
Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinases/metabolism , G1 Phase , Protein Processing, Post-Translational , Amino Acid Motifs , Cell Proliferation , Cyclin D/metabolism , Cyclin H/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinases/genetics , Enzyme Activation , Epistasis, Genetic , HCT116 Cells , Humans , Phosphorylation , Retinoblastoma Protein/metabolism , S Phase , Cyclin-Dependent Kinase-Activating Kinase
8.
PLoS Genet ; 8(8): e1002935, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22927831

ABSTRACT

The cyclin-dependent kinases (CDKs) that promote cell-cycle progression are targets for negative regulation by signals from damaged or unreplicated DNA, but also play active roles in response to DNA lesions. The requirement for activity in the face of DNA damage implies that there are mechanisms to insulate certain CDKs from checkpoint inhibition. It remains difficult, however, to assign precise functions to specific CDKs in protecting genomic integrity. In mammals, Cdk2 is active throughout S and G2 phases, but Cdk2 protein is dispensable for survival, owing to compensation by other CDKs. That plasticity obscured a requirement for Cdk2 activity in proliferation of human cells, which we uncovered by replacement of wild-type Cdk2 with a mutant version sensitized to inhibition by bulky adenine analogs. Here we show that transient, selective inhibition of analog-sensitive (AS) Cdk2 after exposure to ionizing radiation (IR) enhances cell-killing. In extracts supplemented with an ATP analog used preferentially by AS kinases, Cdk2(as) phosphorylated the Nijmegen Breakage Syndrome gene product Nbs1-a component of the conserved Mre11-Rad50-Nbs1 complex required for normal DNA damage repair and checkpoint signaling-dependent on a consensus CDK recognition site at Ser432. In vivo, selective inhibition of Cdk2 delayed and diminished Nbs1-Ser432 phosphorylation during S phase, and mutation of Ser432 to Ala or Asp increased IR-sensitivity. Therefore, by chemical genetics, we uncovered both a non-redundant requirement for Cdk2 activity in response to DNA damage and a specific target of Cdk2 within the DNA repair machinery.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 2/metabolism , DNA Damage/radiation effects , Nuclear Proteins/metabolism , Radiation, Ionizing , Acid Anhydride Hydrolases , Cell Cycle , DNA Repair , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , MRE11 Homologue Protein , Phosphorylation
9.
Cell Cycle ; 11(14): 2600-5, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22732498

ABSTRACT

Progression through the eukaryotic cell division cycle is governed by the activity of cyclin-dependent kinases (CDKs). For a CDK to become active it must (1) bind a positive regulatory subunit (cyclin) and (2) be phosphorylated on its activation (T) loop. In metazoans, multiple CDK catalytic subunits, each with a distinct set of preferred cyclin partners, regulate the cell cycle, but it has been difficult to assign functions to individual CDKs in vivo. Biochemical analyses and experiments with dominant-negative alleles suggested that specific CDK/cyclin complexes regulate different events, but genetic loss of interphase CDKs (Cdk2, -4 and -6), alone or in combination, did not block proliferation of cells in culture. These knockout and knockdown studies suggested redundancy or plasticity built into the CDK network but did not address whether there was true redundancy in normal cells with a full complement of CDKs. Here, we discuss recent work that took a chemical-genetic approach to reveal that the activity of a genetically non-essential CDK, Cdk2, is required for cell proliferation when normal cyclin pairing is maintained. These results have implications for the systems-level organization of the cell cycle, for regulation of the restriction point and G 1/S transition and for efforts to target Cdk2 therapeutically in human cancers.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Drug Evaluation, Preclinical , G1 Phase , Gene Targeting , HCT116 Cells , Humans , S Phase
10.
Proc Natl Acad Sci U S A ; 109(17): E1019-27, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22474407

ABSTRACT

A family of conserved serine/threonine kinases known as cyclin-dependent kinases (CDKs) drives orderly cell cycle progression in mammalian cells. Prior studies have suggested that CDK2 regulates S-phase entry and progression, and frequently shows increased activity in a wide spectrum of human tumors. Genetic KO/knockdown approaches, however, have suggested that lack of CDK2 protein does not prevent cellular proliferation, both during somatic development in mice as well as in human cancer cell lines. Here, we use an alternative, chemical-genetic approach to achieve specific inhibition of CDK2 kinase activity in cells. We directly compare small-molecule inhibition of CDK2 kinase activity with siRNA knockdown and show that small-molecule inhibition results in marked defects in proliferation of nontransformed cells, whereas siRNA knockdown does not, highlighting the differences between these two approaches. In addition, CDK2 inhibition drastically diminishes anchorage-independent growth of human cancer cells and cells transformed with various oncogenes. Our results establish that CDK2 activity is necessary for normal mammalian cell cycle progression and suggest that it might be a useful therapeutic target for treating cancer.


Subject(s)
Cell Transformation, Neoplastic , Cyclin-Dependent Kinase 2/physiology , Oncogenes , Animals , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/genetics , Gene Knockdown Techniques , Humans , Mice , RNA, Small Interfering
11.
Mol Cell ; 42(5): 624-36, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21658603

ABSTRACT

Multiple cyclin-dependent kinases (CDKs) control eukaryotic cell division, but assigning specific functions to individual CDKs remains a challenge. During the mammalian cell cycle, Cdk2 forms active complexes before Cdk1, but lack of Cdk2 protein does not block cell-cycle progression. To detect requirements and define functions for Cdk2 activity in human cells when normal expression levels are preserved, and nonphysiologic compensation by other CDKs is prevented, we replaced the wild-type kinase with a version sensitized to specific inhibition by bulky adenine analogs. The sensitizing mutation also impaired a noncatalytic function of Cdk2 in restricting assembly of cyclin A with Cdk1, but this defect could be corrected by both inhibitory and noninhibitory analogs. This allowed either chemical rescue or selective antagonism of Cdk2 activity in vivo, to uncover a requirement in cell proliferation, and nonredundant, rate-limiting roles in restriction point passage and S phase entry.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 2/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Cell Line , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/genetics , G1 Phase/drug effects , G1 Phase/physiology , Humans , Protein Structure, Tertiary , S Phase/drug effects , S Phase/physiology
12.
F1000 Biol Rep ; 22010 May 11.
Article in English | MEDLINE | ID: mdl-20948801

ABSTRACT

The cell division cycle can be modelled as a series of quantitative thresholds of cyclin-dependent kinase (CDK) activity. DNA synthesis has a lower threshold requirement for CDK than does entry into mitosis, and mitotic exit and re-setting of replication origins occur upon collapse of CDK activity below both thresholds, so that the simple rise and fall of CDK with each cell cycle might suffice to ensure repeated alternation of chromosome duplication and segregation. Recent experimental dissections of mitotic exit, which have both guided and been informed by computational modelling, suggest a more complicated mechanism, in which unidirectional progression is ensured by systems-level control of CDK function and the balance between mitotic CDK and phosphatase activities.

13.
Cell Cycle ; 9(4): 706-14, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20139727

ABSTRACT

Eukaryotic cell division is controlled by the activity of cyclin-dependent kinases (CDKs). Cdk1 and Cdk2, which function at different stages of the mammalian cell cycle, both require cyclin-binding and phosphorylation of the activation (T-) loop for full activity, but differ with respect to the order in which the two steps occur in vivo. To form stable complexes with either of its partners-cyclins A and B-Cdk1 must be phosphorylated on its T-loop, but that phosphorylation in turn depends on the presence of cyclin. Cdk2 can follow a kinetically distinct path to activation in which T-loop phosphorylation precedes cyclin-binding, and thereby out-compete the more abundant Cdk1 for limiting amounts of cyclin A. Mathematical modeling suggests this could be a principal basis for the temporal ordering of CDK activation during S phase, which may dictate the sequence in which replication origins fire. Still to be determined are how: (1) the activation machinery discriminates between closely related CDKs, and (2) coordination of the cell cycle is affected when this mechanism of pathway insulation breaks down.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin A/metabolism , Cyclin B/metabolism , DNA Replication , Models, Theoretical , Phosphorylation , Protein Interaction Domains and Motifs , S Phase
14.
Mol Cell ; 32(5): 662-72, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-19061641

ABSTRACT

In metazoans, different cyclin-dependent kinases (CDKs) bind preferred cyclin partners to coordinate cell division. Here, we investigate these preferences in human cells and show that cyclin A assembles with Cdk1 only after complex formation with Cdk2 reaches a plateau during late S and G2 phases. To understand the basis for Cdk2's competitive advantage, despite Cdk1's greater abundance, we dissect their activation pathways by chemical genetics. Cdk1 and Cdk2 are activated by kinetically distinct mechanisms, even though they share the same CDK-activating kinase (CAK), Cdk7. We recapitulate cyclin A's selectivity for Cdk2 in extracts and override it with a yeast CAK that phosphorylates monomeric Cdk1, redirecting Cdk1 into a pathway normally restricted to Cdk2. Conversely, upon Cdk7 inhibition in vivo, cyclin B, which normally binds Cdk1 nearly exclusively, is diverted to Cdk2. Therefore, differential ordering of common activation steps promotes CDK-cyclin specificity, with Cdk7 acting catalytically to enforce fidelity.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclins/metabolism , Animals , Cell Cycle , Cell Extracts , Cyclin-Dependent Kinases/metabolism , Enzyme Activation , Enzyme Stability , HCT116 Cells , Humans , Models, Biological , Phosphates/metabolism , Phosphorylation , Phosphothreonine/metabolism , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Cyclin-Dependent Kinase-Activating Kinase
15.
Mol Cell ; 25(6): 839-50, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17386261

ABSTRACT

Cell division is controlled by cyclin-dependent kinases (CDKs). In metazoans, S phase onset coincides with activation of Cdk2, whereas Cdk1 triggers mitosis. Both Cdk1 and -2 require cyclin binding and T loop phosphorylation for full activity. The only known CDK-activating kinase (CAK) in metazoans is Cdk7, which is also part of the transcription machinery. To test the requirements for Cdk7 in vivo, we replaced wild-type Cdk7 with a version sensitive to bulky ATP analogs in human cancer cells. Selective inhibition of Cdk7 in G1 prevents activation (but not formation) of Cdk2/cyclin complexes and delays S phase. Inhibiting Cdk7 in G2 blocks entry to mitosis and disrupts Cdk1/cyclin B complex assembly, indicating that the two steps of Cdk1 activation-cyclin binding and T loop phosphorylation-are mutually dependent. Therefore, by combining chemical genetics and homologous gene replacement in somatic cells, we reveal different modes of CDK activation by Cdk7 at two distinct execution points in the cell cycle.


Subject(s)
CDC2 Protein Kinase/genetics , Cyclin B/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/metabolism , Cell Cycle , Cell Division , Cell Survival , Enzyme Activation , Humans , Mitosis , S Phase , Cyclin-Dependent Kinase-Activating Kinase
16.
Opt Express ; 15(26): 18220-35, 2007 Dec 24.
Article in English | MEDLINE | ID: mdl-19551120

ABSTRACT

Fluorescence lifetime imaging microscopy (FLIM) provides a promising, robust method of detecting molecular interaction not not nots in vivo via fluorescence/Förster resonance energy transfer (FRET), by monitoring the variation of donor fluorescence lifetime, which is insensitive to many artifacts influencing convential intensity-based measurements, e.g. fluorophore concentration, photobleaching, and spectral bleed-through. As proof of principle, we demonstrate the capability of a novel picosecond-resolution FLIM system to detect molecular interactions in a well-established FRET assay. We then apply the FLIM system to detect the molecular interaction of a transforming oncogene RhoC with a binding partner RhoGDIgamma in vivo, which is critical to understand and interfere with Rho signaling for cancer therapeutics.


Subject(s)
Fluorescence Resonance Energy Transfer/instrumentation , Image Enhancement/instrumentation , Microscopy, Fluorescence/instrumentation , Protein Interaction Mapping/instrumentation , Computer-Aided Design , Equipment Design , Equipment Failure Analysis , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...