Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cancer Discov ; 12(2): 356-371, 2022 02.
Article in English | MEDLINE | ID: mdl-34544752

ABSTRACT

Cyclin-dependent kinases 4 and 6 (CDK4/6) represent a major therapeutic vulnerability for breast cancer. The kinases are clinically targeted via ATP competitive inhibitors (CDK4/6i); however, drug resistance commonly emerges over time. To understand CDK4/6i resistance, we surveyed over 1,300 breast cancers and identified several genetic alterations (e.g., FAT1, PTEN, or ARID1A loss) converging on upregulation of CDK6. Mechanistically, we demonstrate CDK6 causes resistance by inducing and binding CDK inhibitor INK4 proteins (e.g., p18INK4C). In vitro binding and kinase assays together with physical modeling reveal that the p18INK4C-cyclin D-CDK6 complex occludes CDK4/6i binding while only weakly suppressing ATP binding. Suppression of INK4 expression or its binding to CDK6 restores CDK4/6i sensitivity. To overcome this constraint, we developed bifunctional degraders conjugating palbociclib with E3 ligands. Two resulting lead compounds potently degraded CDK4/6, leading to substantial antitumor effects in vivo, demonstrating the promising therapeutic potential for retargeting CDK4/6 despite CDK4/6i resistance. SIGNIFICANCE: CDK4/6 kinase activation represents a common mechanism by which oncogenic signaling induces proliferation and is potentially targetable by ATP competitive inhibitors. We identify a CDK6-INK4 complex that is resilient to current-generation inhibitors and develop a new strategy for more effective inhibition of CDK4/6 kinases.This article is highlighted in the In This Issue feature, p. 275.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Drug Resistance, Neoplasm , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor/drug effects , Cyclin-Dependent Kinase Inhibitor Proteins/administration & dosage , Cyclin-Dependent Kinase Inhibitor Proteins/therapeutic use , Female , Humans , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Tumor Suppressor Proteins/metabolism
2.
Methods Mol Biol ; 2141: 429-445, 2020.
Article in English | MEDLINE | ID: mdl-32696370

ABSTRACT

The level of compaction of an intrinsically disordered protein may affect both its physical and biological properties, and can be probed via different types of biophysical experiments. Small-angle X-ray scattering (SAXS) probe the radius of gyration (Rg) whereas pulsed-field-gradient nuclear magnetic resonance (NMR) diffusion, fluorescence correlation spectroscopy, and dynamic light scattering experiments can be used to determine the hydrodynamic radius (Rh). Here we show how to calculate Rg and Rh from a computationally generated conformational ensemble of an intrinsically disordered protein. We further describe how to use a Bayesian/Maximum Entropy procedure to integrate data from SAXS and NMR diffusion experiments, so as to derive conformational ensembles in agreement with those experiments.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Protein Conformation , Scattering, Small Angle , X-Ray Diffraction/methods , Amino Acid Sequence , Bayes Theorem , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Electrophoresis, Gel, Pulsed-Field , Entropy , F-Box Proteins/metabolism , Hydrodynamics , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Imaging , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular/methods , Phosphorylation , Protein Folding , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Spectrometry, Fluorescence , Static Electricity , Ubiquitin-Protein Ligases/metabolism
3.
Mol Inform ; 39(5): e1900126, 2020 05.
Article in English | MEDLINE | ID: mdl-31943821

ABSTRACT

In this study, we evaluated the effect of structural descriptors on the in silico design of bioactive compounds. The authors have proposed a molecular design technique for designing new bioactive compounds. In this approach, known fragments are combined to generate new structures, which are evolved to increase the similarity to a known active compound. We generated the structure of CDK2 inhibitors using four descriptors (three binary fingerprints and a numerical vector) to evaluate the effect of descriptors on the molecular design. Subsequently, the physicochemical properties of the generated compounds were compared and evaluated from a similarity viewpoint. As a result, it was clarified that better structures can be generated by using descriptors consisting of numerical vectors rather than binary fingerprints. Moreover, the compound generated using the numerical vector or a long-bit fingerprint resulted in favorable docking scores. Although binary fingerprints such as MACCS are widely used in this field, this result shows that it is important to use numeric vectors, or at least to use long-bit fingerprints, to design drug-like CDK2 inhibitors by the similarity-based structure generation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinases/chemistry , Molecular Docking Simulation/methods , Algorithms , Binding Sites , Computer Simulation , Cyclin-Dependent Kinases/antagonists & inhibitors , Databases, Chemical , Drug Design , Evolution, Molecular , Models, Chemical , Molecular Structure , Molecular Weight , Mutation , Structure-Activity Relationship
4.
Sci Rep ; 9(1): 1575, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733475

ABSTRACT

The mammalian CIP/KIP family proteins are intrinsically disordered proteins (IDPs) that can regulate various cellular processes. However, many reports have shown that IDPs generally evolve more rapidly than ordered proteins. Here, to elucidate the functional adaptability of CIP/KIP proteins in vertebrate, we analysed the rates of evolution in relation to their structural and sequence properties and predicted the post-translational modification based on the sequence data. The results showed that CIP/KIP proteins generally could maintain their function through evolution in the vertebrate. Basically, the disordered region that acts as a flexible linker or spacer has a conserved propensity for structural disorder and a persistent, fast rate of amino acid substitution, which could result in a significantly faster rate of evolution compared to the ordered proteins. Describing the pattern of structural order-disorder evolution, this study may give an insight into the well-known characteristics of IDPs in the evolution of CIP/KIP proteins.


Subject(s)
Calcium-Binding Proteins/chemistry , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Intrinsically Disordered Proteins/chemistry , Amino Acid Sequence , Calcium-Binding Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Evolution, Molecular , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Models, Molecular , Phosphorylation , Phylogeny , Protein Conformation , Structure-Activity Relationship
5.
Plant Physiol ; 177(4): 1569-1579, 2018 08.
Article in English | MEDLINE | ID: mdl-29903833

ABSTRACT

Endoreplication, a modified cell cycle in which DNA is replicated without subsequent cell division, plays an important but poorly understood role in plant growth and in plant responses to biotic and abiotic stress. The Arabidopsis (Arabidopsis thaliana) SIAMESE (SIM) gene encodes the first identified member of the SIAMESE-RELATED (SMR) family of cyclin-dependent kinase inhibitors. SIM controls endoreplication during trichome development, and sim mutant trichomes divide several times instead of endoreplicating their DNA. The SMR family is defined by several short linear amino acid sequence motifs of largely unknown function, and family members have little sequence similarity to any known protein functional domains. Here, we investigated the roles of the conserved motifs in SIM site-directed Arabidopsis mutants using several functional assays. We identified a potential cyclin-dependent kinase (CDK)-binding site, which bears no resemblance to other known CDK interaction motifs. We also identified a potential site of phosphorylation and two redundant nuclear localization sequences. Surprisingly, the only motif with similarity to the other family of plant CDK inhibitors, the INHIBITOR/INTERACTOR OF CDC2 KINASE/KIP-RELATED PROTEIN proteins, is not required for SIM function in vivo. Because even highly divergent members of the SMR family are able to replace SIM function in Arabidopsis trichomes, it is likely that the results obtained here for SIM will apply to other members of this plant-specific family of CDK inhibitors.


Subject(s)
Amino Acid Motifs , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Cell Cycle Proteins/genetics , Conserved Sequence , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Mutation , Nuclear Localization Signals , Phosphorylation , Plants, Genetically Modified , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational
6.
Biochim Biophys Acta Gen Subj ; 1862(7): 1556-1564, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29621630

ABSTRACT

BACKGROUND: Protein-nanoparticle (NP) interactions dictate properties of nanoconjugates relevant to bionanotechnology. Non-covalent adsorption generates a protein corona (PC) formed by an inner and an outer layer, the hard and soft corona (HC, SC). Intrinsically disordered proteins (IDPs) exist in solution as conformational ensembles, whose response to the presence of NPs is not known. METHODS: Three IDPs (α-casein, Sic1 and α-synuclein) and lysozyme are compared, describing conformational properties inside HC on silica NPs by circular dichroism (CD) and Fourier-transform infrared (FTIR) spectroscopy. RESULTS: IDPs inside HC are largely unstructured, but display small, protein-specific conformational changes. A minor increase in helical content is observed for α-casein and α-synuclein, reminiscent of membrane effects on α-synuclein. Frozen in their largely disordered conformation, bound proteins do not undergo folding induced by dehydration, as they do in their free forms. While HC thickness approaches the hydrodynamic diameter of the protein in solution for lysozyme, it is much below the respective values for IDPs. NPs boost α-synuclein aggregation kinetics in a dose-dependent manner. CONCLUSIONS: IDPs maintain structural disorder inside HC, experiencing minor, protein-specific, induced folding and stabilization against further conformational transitions, such as formation of intermolecular beta-sheets upon dehydration. The HC is formed by a single layer of protein molecules. SC likely plays a key role stabilizing amyloidogenic α-synuclein conformers. GENERAL SIGNIFICANCE: Protein-NP interactions can mimic those with macromolecular partners, allowing dissection of contributing factors by rational design of NP surfaces. Application of NPs in vivo should be carefully tested for amyloidogenic potential.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Nanoparticles , Protein Conformation , Protein Corona/chemistry , Animals , Caseins/chemistry , Cattle , Chick Embryo , Circular Dichroism , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Muramidase/chemistry , Protein Binding , Saccharomyces cerevisiae Proteins/chemistry , Silicon Dioxide , Spectroscopy, Fourier Transform Infrared , alpha-Synuclein/chemistry
7.
Int J Biochem Cell Biol ; 91(Pt B): 98-101, 2017 10.
Article in English | MEDLINE | ID: mdl-28504190

ABSTRACT

The cyclin-dependent kinase inhibitor-3 (CDKN3) gene encodes a dual-specificity protein tyrosine phosphatase that dephosphorylates CDK1/CDK2 and other proteins. CDKN3 is often overexpressed in human cancer, and this overexpression correlates with reduced survival in several types of cancer. CDKN3 transcript variants and mutations have also been reported. The mechanism of CDKN3 overexpression and the role of CDKN3 transcript variants in human cancer are not entirely clear. Here, we review the literature and provide additional data to assess the correlation of CDKN3 expression with patient survival. Besides the full-length CDKN3 encoding transcript and a major transcript that skips exon 2 express in normal and cancer cells, minor aberrant transcript variants have been reported. Aberrant CDKN3 transcripts were postulated to encode dominant-negative inhibitors of CDKN3 as an explanation for overexpression of the perceived tumor suppressor gene in human cancer. However, while CDKN3 is often overexpressed in human cancer, aberrant CDKN3 transcripts occur infrequently and at lower levels. CDKN3 mutations and copy number alternation are rare in human cancer, implying that neither loss of CDKN3 activity nor constitutive gain of CDKN3 expression offer an advantage to tumorigenesis. Recently, it was found that CDKN3 transcript and protein levels fluctuate during the cell cycle, peaking in mitosis. Given that rapidly growing tumors have more mitotic cells, the high level of mitotic CDKN3 expression is the most plausible mechanism of frequent CDKN3 overexpression in human cancer. This finding clarifies the mechanism of CDKN3 overexpression in human cancer and questions the view of CDKN3 as a tumor suppressor.


Subject(s)
Alternative Splicing , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/enzymology , Neoplasms/genetics , Amino Acid Sequence , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Dual-Specificity Phosphatases/chemistry , Humans , Mitosis/genetics , Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis
8.
Sci Rep ; 7: 43818, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262730

ABSTRACT

Inhibitors of cyclin-dependent kinases (ICKs) are key regulators of cyclin-dependent kinase activities and cell division. Herein, we identified eight ICKs in maize, which we named Zeama;ICKs (ZmICKs). Primary sequencing and phylogenetic analyses were used to divide the ZmICK family into two classes: group B and group C. Subcellular localization analysis of ZmICK:enhanced green fluorescent protein (eGFP) fusion constructs in tobacco leaf cells indicated that ZmICKs are principally nuclear. Co-localization analysis of the ZmICKs and maize A-type cyclin-dependent kinase (ZmCDKA) was also performed using enhanced green fluorescent protein (eGFP) and red fluorescent protein (RFP) fusion constructs. The ZmICKs and ZmCDKA co-localized in the nucleus. Semi-quantitative RT-PCR analysis of the ZmICKs showed that they were expressed at different levels in all tissues examined and shared similar expression patterns with cell cycle-related genes. Yeast two-hybrid and bimolecular fluorescence complementation assays showed that ZmICK1, ZmICK2, ZmICK3, and ZmICK4 interact with ZmCDKA1 and ZmCDKA3. Interestingly, ZmICK7 interacts with D-type cyclins. Transformed and expressed ZmCDKA1 and ZmICKs together in fission yeast revealed that ZmICK1, ZmICK3, and ZmICK4 can affect ZmCDKA1 function. Moreover, the C-group of ZmICKs could interact with ZmCDKA1 directly and affect ZmCDKA1 function, suggesting that C-group ZmICKs are important for cell division regulation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Gene Expression Regulation, Plant , Multigene Family , Plant Proteins/genetics , Zea mays/genetics , Cell Cycle/genetics , Cell Division/genetics , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Phylogeny , Plant Proteins/classification , Plant Proteins/metabolism , Plants, Genetically Modified , Protein Binding , Zea mays/metabolism
9.
Nat Commun ; 8: 13943, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045046

ABSTRACT

The ubiquitin ligase SCFCdc4 mediates phosphorylation-dependent elimination of numerous substrates by binding one or more Cdc4 phosphodegrons (CPDs). Methyl-based NMR analysis of the Cdc4 WD40 domain demonstrates that Cyclin E, Sic1 and Ash1 degrons have variable effects on the primary Cdc4WD40 binding pocket. Unexpectedly, a Sic1-derived multi-CPD substrate (pSic1) perturbs methyls around a previously documented allosteric binding site for the chemical inhibitor SCF-I2. NMR cross-saturation experiments confirm direct contact between pSic1 and the allosteric pocket. Phosphopeptide affinity measurements reveal negative allosteric communication between the primary CPD and allosteric pockets. Mathematical modelling indicates that the allosteric pocket may enhance ultrasensitivity by tethering pSic1 to Cdc4. These results suggest negative allosteric interaction between two distinct binding pockets on the Cdc4WD40 domain may facilitate dynamic exchange of multiple CPD sites to confer ultrasensitive dependence on substrate phosphorylation.


Subject(s)
Cyclin E/chemistry , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Gene Expression Regulation, Fungal , Repressor Proteins/chemistry , SKP Cullin F-Box Protein Ligases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Allosteric Regulation , Allosteric Site , Binding Sites , Cloning, Molecular , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Kinetics , Models, Molecular , Phosphopeptides , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Thermodynamics
10.
J Am Soc Mass Spectrom ; 28(1): 21-28, 2017 01.
Article in English | MEDLINE | ID: mdl-27730522

ABSTRACT

Native mass spectrometry (MS) has become a central tool of structural proteomics, but its applicability to the peculiar class of intrinsically disordered proteins (IDPs) is still object of debate. IDPs lack an ordered tridimensional structure and are characterized by high conformational plasticity. Since they represent valuable targets for cancer and neurodegeneration research, there is an urgent need of methodological advances for description of the conformational ensembles populated by these proteins in solution. However, structural rearrangements during electrospray-ionization (ESI) or after the transfer to the gas phase could affect data obtained by native ESI-MS. In particular, charge-state distributions (CSDs) are affected by protein conformation inside ESI droplets, while ion mobility (IM) reflects protein conformation in the gas phase. This review focuses on the available evidence relating IDP solution ensembles with CSDs, trying to summarize cases of apparent consistency or discrepancy. The protein-specificity of ionization patterns and their responses to ligands and buffer conditions suggests that CSDs are imprinted to protein structural features also in the case of IDPs. Nevertheless, it seems that these proteins are more easily affected by electrospray conditions, leading in some cases to rearrangements of the conformational ensembles. Graphical Abstract ᅟ.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Humans , Models, Molecular , Protein Conformation , Protein Multimerization , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Static Electricity , Tumor Suppressor Protein p53/chemistry
11.
Methods Mol Biol ; 1336: 59-66, 2016.
Article in English | MEDLINE | ID: mdl-26231708

ABSTRACT

The homology of the inhibitor binding site regions on the surface of cyclin-dependent kinases (CDKs) makes actual CDK inhibitors unable to bind specifically to their molecular targets. Most of them are ATP competitive inhibitors with low specificity that also affect the phosphorylation mechanisms of other nontarget kinases giving rise to harmful side effects. So, the search of specific and potent inhibitors able to bind to the desired CDK target is still a pending issue. Structure based drug design minimized the erroneous binding and increased the affinity of the inhibitor interaction. In the case of CDKs their activation and regulation mechanisms mainly depend on protein-protein interactions (PPIs). The design of drugs targeting these PPIs makes feasible and promising towards the discovery of new and specific CDK inhibitors. Development of peptide inhibitors for a target protein is an emerging approach in computer aided drug designing. This chapter describes in detail methodology for use of the VitAL-Viterbi algorithm for de novo peptide design of CDK2 inhibitors.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/chemistry , Protein Interaction Mapping , Algorithms , Amino Acids/chemistry , Binding Sites , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Databases, Protein , Drug Design , Humans , Models, Molecular , Models, Statistical , Peptides/chemistry , Phosphorylation , Protein Structure, Secondary , Software , Structure-Activity Relationship , Thermodynamics
12.
Methods Mol Biol ; 1336: 47-58, 2016.
Article in English | MEDLINE | ID: mdl-26231707

ABSTRACT

Cyclin-dependent kinases (CDKs) are core components of the cell cycle machinery that govern the transition between phases during cell cycle progression. Abnormalities in CDKs activity and regulation are common features of cancer, making CDK family members attractive targets for the development of anticancer drugs. One of the main bottlenecks hampering the development of drugs for kinase is the difficulty to attain selectivity. A huge variety of small molecules have been reported as CDK inhibitors, as potential anticancer agents, but none of these has been approved for commercial use. Computer-based molecular design supports drug discovery by suggesting novel new chemotypes and compound modifications for lead candidate optimization. One of the methods known as de novo ligand design technique has emerged as a complementary approach to high-throughput screening. Several automated de novo software programs have been written, which automatically design novel structures to perfectly fit in known binding site. The de novo design supports drug discovery assignments by generating novel pharmaceutically active agents with desired properties in a cost as well as time efficient approach. This chapter describes procedure and an overview of computer-based molecular de novo design methods on a conceptual level with successful examples of CDKs inhibitors.


Subject(s)
Adenosine Triphosphate/chemistry , Crystallography, X-Ray/methods , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Algorithms , Antineoplastic Agents/chemistry , Binding Sites , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Databases, Protein , Drug Design , Humans , Ligands , Molecular Biology/methods , Nucleic Acid Conformation , Protein Binding , Software
13.
Methods Mol Biol ; 1336: 85-93, 2016.
Article in English | MEDLINE | ID: mdl-26231710

ABSTRACT

Cell synchronization techniques have been used for the studies of mechanisms involved in cell cycle regulation. Synchronization involves the enrichment of subpopulations of cells in specific stages of the cell cycle. These subpopulations are then used to study regulatory mechanisms of the cell cycle such as DNA synthesis, gene expression, protein synthesis, protein phosphorylation, protein degradation, and development of new drugs (e.g., CDK inhibitors). Here, we describe several protocols for synchronization of cells from different phases of the cell cycle. We also describe protocols for determining cell viability and mitotic index and for validating the synchrony of the cells by flow cytometry.


Subject(s)
Cell Culture Techniques/methods , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Animals , Aphidicolin/chemistry , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , DNA/chemistry , DNA Replication , Flow Cytometry , HeLa Cells , Humans , Mice , Mitosis , Mitotic Index , NIH 3T3 Cells , Nocodazole/chemistry , Thymidine/chemistry , Time Factors , Trypan Blue/chemistry
14.
Methods Mol Biol ; 1336: 111-21, 2016.
Article in English | MEDLINE | ID: mdl-26231712

ABSTRACT

CDK inhibitors have been used to induce protection in various experimental models. Kidney ischemia-reperfusion (I/R) is a form of acute kidney injury resulting in a cascade of cellular events prompting rapid cellular damage and suppression of kidney function. I/R injury, an inevitable impairment during renal transplant surgery, remains one of the major causes of acute kidney injury and represents the most prominent factor leading to delayed graft function after transplantation. Understanding the molecular events responsible for tubule damage and recovery would help to develop new strategies for organ preservation. This chapter describes procedures to study the effect of CDK inhibitors in the cellular I/R model developed from an epithelial cell line deriving from pig kidney proximal tubule cells (LLC-PK1). We briefly describe methods for determining the protective effect of CDK inhibitors such as activation of caspase 3/7, western blot analysis, gene silencing, and immunoprecipitation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinases/antagonists & inhibitors , Kidney/pathology , Reperfusion Injury/pathology , Acute Kidney Injury , Animals , Blotting, Western , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Epithelial Cells/metabolism , Gene Silencing , Immunoprecipitation , Kidney Transplantation , Kidney Tubules, Proximal/cytology , Molecular Biology/methods , RNA, Small Interfering/metabolism , Swine
15.
Methods Mol Biol ; 1336: 167-78, 2016.
Article in English | MEDLINE | ID: mdl-26231716

ABSTRACT

The cell cycle of eukaryotic cells is regulated by a family of protein kinases called cyclin-dependent kinases (Cdks). We have reported the identification and biological characterization of a highly potent, small-molecule pan-Cdk inhibitor, which inhibited Cdk1, 2, 4, 5, 6, and 9 with equal potency in the nM range. This compound inhibited multiple events in the cell cycle and induced cell death in human cancer cell lines as well as in peripheral blood or purified resting lymphocytes ex vivo. We describe the materials and methods to determine antitumor efficacy in vivo xenograft models. Pharmacodynamic marker assays that have been performed using tumors and normal tissues are explained. Moreover, we briefly describe methods for determining the effects of chemical Cdk inhibitors on peripheral blood cells or lymphocytes ex vivo.


Subject(s)
Cell Death/drug effects , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Cycle , Drug Design , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Immunohistochemistry , Lymphocytes/cytology , Lymphocytes/drug effects , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Rats, Nude , Xenograft Model Antitumor Assays
16.
Methods Mol Biol ; 1336: 211-23, 2016.
Article in English | MEDLINE | ID: mdl-26231718

ABSTRACT

Cyclin-dependent kinases (CDKs) regulate cell cycle progression, and some of them are also involved in the control of cellular transcription. Dysregulation of these critical cellular processes, due to the aberrant expression of some of these proteins, is common in many neoplastic malignancies. Consequently, the development of chemical compounds capable of inhibiting the biological activity of CDKs represents an attractive strategy in the anticancer area. CDK inhibition can trigger apoptosis and could be particularly useful in hematological malignancies, which are more sensitive to inhibition of cell cycle and apoptosis induction. Over the last few years, a number of pharmacological inhibitors of CDKs (CDKIs) belonging to different chemical families have been developed, and some of them have been tested in clinical trials. Given the complexity of the role of CDKs in cell functioning, it would be desirable to develop new tools that could facilitate a better understanding of the new insights into CDK functions and the mode-of-actions of CDKIs. In this context, this chapter describes an experimental approach to evaluate the metabolic consequences of CDKIs at the cellular level based on metabolomics by NMR. More specifically, a description of a strategy to characterize the biochemical effects of CDKIs acting on mammalian cells is provided, including protocols for the extraction of hydrophilic and lipophilic metabolites, the acquisition of 1D and 2D metabolomic Nuclear Magnetic Resonance (NMR) experiments, the identification and quantification of metabolites, and the annotation of the results in the context of biochemical pathways.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinases/antagonists & inhibitors , Metabolomics/methods , Protein Kinase Inhibitors/chemistry , Animals , Apoptosis/drug effects , Cell Adhesion , Cell Culture Techniques , Cell Cycle/drug effects , Cell Division , Clinical Trials as Topic , Humans , Magnetic Resonance Spectroscopy , Neoplasms/drug therapy
17.
Br J Cancer ; 113(12): 1735-43, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26554648

ABSTRACT

BACKGROUND: The cyclin-dependent kinase inhibitor 3 (CDKN3) has been perceived as a tumour suppressor. Paradoxically, CDKN3 is often overexpressed in human cancer. It was unclear if CDKN3 overexpression is linked to alternative splicing variants or mutations that produce dominant-negative CDKN3. METHODS: We analysed CDKN3 expression and its association with patient survival in three cohorts of lung adenocarcinoma. We also examined CDKN3 mutations in the Cancer Genome Atlas (TCGA) and the Moffitt Cancer Center's Total Cancer Care (TCC) projects. CDKN3 transcripts were further analysed in a panel of cell lines and lung adenocarcinoma tissues. CDKN3 mRNA and protein levels in different cell cycle phases were examined. RESULTS: CDKN3 is overexpressed in non small cell lung cancer. High CDKN3 expression is associated with poor overall survival in lung adenocarcinoma. Two CDKN3 transcripts were detected in all samples. These CDKN3 transcripts represent the full length CDKN3 mRNA and a normal transcript lacking exon 2, which encodes an out of frame 23-amino acid peptide with little homology to CDKN3. CDKN3 mutations were found to be very rare. CDKN3 mRNA and protein were elevated during the mitosis phase of cell cycle. CONCLUSIONS: CDKN3 overexpression is prognostic of poor overall survival in lung adenocarcinoma. CDKN3 overexpression in lung adenocarcinoma is not attributed to alternative splicing or mutation but is likely due to increased mitotic activity, arguing against CDKN3 as a tumour suppressor.


Subject(s)
Adenocarcinoma/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dual-Specificity Phosphatases/genetics , Lung Neoplasms/genetics , RNA, Messenger/genetics , Survival Analysis , Amino Acid Sequence , Cohort Studies , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Dual-Specificity Phosphatases/chemistry , Humans , Molecular Sequence Data
18.
Chem Biol Drug Des ; 86(6): 1528-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26198005

ABSTRACT

Selective inhibitors of kinases that regulate the cell cycle, such as cyclin-dependent kinases (CDKs) and aurora kinases, could potentially become powerful tools for the treatment of cancer. We prepared and studied a series of 3,5,7-trisubstituted pyrazolo[4,3-d]pyrimidines, a new CDK inhibitor scaffold, to assess their CDK2 inhibitory and antiproliferative activities. A new compound, 2i, which preferentially inhibits CDK2, CDK5, and aurora A was identified. Both biochemical and cellular assays indicated that treatment with compound 2i caused the downregulation of cyclins A and B, the dephosphorylation of histone H3 at Ser10, and the induction of mitochondrial apoptosis in the HCT-116 colon cancer cell line. It also reduced migration as well as tube and lamellipodia formation in human endothelial cells. The kinase inhibitory profile of compound 2i suggests that its anti-angiogenic activity is linked to CDK5 inhibition. This dual mode of action involving apoptosis induction in cancer cells and the blocking of angiogenesis-like activity in endothelial cells offers possible therapeutic potential.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor Proteins/chemical synthesis , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Cyclin-Dependent Kinase Inhibitor Proteins/pharmacology , Drug Design , Drug Screening Assays, Antitumor , HCT116 Cells , Human Umbilical Vein Endothelial Cells , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Structure-Activity Relationship
19.
J Phys Chem B ; 118(15): 4088-97, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24673507

ABSTRACT

The yeast cyclin-dependent kinase inhibitor Sic1 is a disordered protein that, upon multisite phosphorylation, forms a dynamic complex with the Cdc4 subunit of an SCF ubiquitin ligase. To understand the multisite phosphorylation dependence of the Sic1:Cdc4 interaction, which ultimately leads to a sharp cell cycle transition, the conformational properties of the disordered Sic1 N-terminal targeting region were studied using single-molecule fluorescence spectroscopy. Multiple conformational populations with different sensitivities to charge screening were identified by performing experiments in nondenaturing salts and ionic denaturants. Both the end-to-end distance and the hydrodynamic radius decrease monotonically with increasing the salt concentration, and a rollover of the chain dimensions in high denaturant conditions is observed. The data were fit to the polyelectrolyte binding-screening model, yielding parameters such as the excluded volume of the uncharged chain and the binding constant to denaturant. An overall scaling factor of ∼1.2 was needed for fitting the data, which implies that Sic1 cannot be approximated by a random Gaussian chain. Fluorescence correlation spectroscopy reveals Sic1 structure fluctuations occurring on both fast (10-100 ns) and slow (∼10 ms) time scales, with the fast phase absent in low salt solutions. The results of this study provide direct evidence that long-range intrachain electrostatic repulsions are a significant factor for the conformational landscape of Sic1, and support the role of electrostatics in determining the overall shape and hydrodynamic properties of intrinsically disordered proteins.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Hydrodynamics , Saccharomyces cerevisiae Proteins/chemistry , Protein Conformation , Static Electricity
20.
Angew Chem Int Ed Engl ; 52(36): 9399-408, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-23934828

ABSTRACT

Innovation has frequently been described as the key to drug discovery. However, in the daily routine, medicinal chemists often tend to stick to the functional groups and structural elements they know and love. Blockbuster cancer drug Velcade (bortezomib), for example, was rejected by more than 50 companies, supposedly because of its unusual boronic acid function (as often repeated: "only a moron would put boron in a drug!"). Similarly, in the discovery process of the pan-CDK inhibitor BAY 1000394, the unconventional proposal to introduce a sulfoximine group into the lead series also led to sneers and raised eyebrows, since sulfoximines have seldom been used in medicinal chemistry. However, it was the introduction of the sulfoximine group that finally allowed the fundamental issues of the project to be overcome, culminating in the identification of the clinical sulfoximine pan-CDK inhibitor BAY 1000394. This Minireview provides an overview of a widely neglected opportunity in medicinal chemistry--the sulfoximine group.


Subject(s)
Chemistry, Pharmaceutical/trends , Methionine Sulfoximine/chemistry , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Humans , Molecular Structure , Pyrimidines/chemistry , Sulfoxides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...