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










Publication year range
1.
Cancer Sci ; 113(1): 132-144, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34668620

ABSTRACT

In solid tumors, cancer cells have devised multiple approaches to survival and proliferate in response to glucose starvation that is often observed in solid tumor microenvironments. However, the precise mechanisms are far less known. Herein, we report that glucose deprivation activates 90-kDa ribosomal S6 kinase (p90 RSK), a highly conserved Ser/Thr kinase, and activated p90 RSK promotes cancer cell survival. Mechanistically, activated p90 RSK by glucose deprivation phosphorylates checkpoint kinase 1 (CHK1), a key transducer in checkpoint signaling pathways, at Ser280 and triggers CHK1 ubiquitination mediated by SCFß-TrCP ubiquitin ligase and proteasomal degradation, subsequently suppressing cancer cell apoptosis induced by glucose deprivation. Importantly, we identified an inverse correlation between p90 RSK activity and CHK1 levels within the solid tumor mass, with lower levels of CHK1 and higher activity of p90 RSK in the center of the tumor where low glucose concentrations are often observed. Thus, our study indicates that p90 RSK promotes CHK1 phosphorylation at Ser280 and its subsequent degradation, which allows cancer cells to escape from checkpoint signals under the stress of glucose deprivation, leading to cell survival and thus contributing to tumorigenesis.


Subject(s)
Checkpoint Kinase 1/metabolism , Glucose/deficiency , Neoplasms/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Cell Line, Tumor , Cell Survival , Checkpoint Kinase 1/chemistry , Enzyme Activation , HEK293 Cells , Humans , Mice , Phosphorylation , Proteolysis/drug effects , Pteridines/pharmacology , Ubiquitination/drug effects
2.
J Med Chem ; 64(14): 10312-10332, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34184879

ABSTRACT

Inhibitors of leucine-rich repeat kinase 2 (LRRK2) and mutants, such as G2019S, have potential utility in Parkinson's disease treatment. Fragment hit-derived pyrrolo[2,3-d]pyrimidines underwent optimization using X-ray structures of LRRK2 kinase domain surrogates, based on checkpoint kinase 1 (CHK1) and a CHK1 10-point mutant. (2R)-2-Methylpyrrolidin-1-yl derivative 18 (LRRK2 G2019S cKi 0.7 nM, LE 0.66) was identified, with increased potency consistent with an X-ray structure of 18/CHK1 10-pt. mutant showing the 2-methyl substituent proximal to Ala147 (Ala2016 in LRRK2). Further structure-guided elaboration of 18 gave the 2-[(1,3-dimethyl-1H-pyrazol-4-yl)amino] derivative 32. Optimization of 32 afforded diastereomeric oxolan-3-yl derivatives 44 and 45, which demonstrated a favorable in vitro PK profile, although they displayed species disconnects in the in vivo PK profile, and a propensity for P-gp- and/or BCRP-mediated efflux in a mouse model. Compounds 44 and 45 demonstrated high potency and exquisite selectivity for LRRK2 and utility as chemical probes for the study of LRRK2 inhibition.


Subject(s)
Checkpoint Kinase 1/chemistry , Drug Design , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Checkpoint Kinase 1/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship
3.
Structure ; 29(6): 531-539.e3, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33789090

ABSTRACT

CHK1 is a protein kinase that functions downstream of activated ATR to phosphorylate multiple targets as part of intra-S and G2/M DNA damage checkpoints. Its role in allowing cells to survive replicative stress has made it an important target for anti-cancer drug discovery. Activation of CHK1 by ATR depends on their mutual interaction with CLASPIN, a natively unstructured protein that interacts with CHK1 through a cluster of phosphorylation sites in its C-terminal half. We have now determined the crystal structure of the kinase domain of CHK1 bound to a high-affinity motif from CLASPIN. Our data show that CLASPIN engages a conserved site on CHK1 adjacent to the substrate-binding cleft, involved in phosphate sensing in other kinases. The CLASPIN motif is not phosphorylated by CHK1, nor does it affect phosphorylation of a CDC25 substrate peptide, suggesting that it functions purely as a scaffold for CHK1 activation by ATR.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Animals , Binding Sites , Checkpoint Kinase 1/genetics , Crystallography, X-Ray , Humans , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Sf9 Cells
4.
Mol Cell ; 81(6): 1231-1245.e8, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33503405

ABSTRACT

ATR checkpoint signaling is crucial for cellular responses to DNA replication impediments. Using an optogenetic platform, we show that TopBP1, the main activator of ATR, self-assembles extensively to yield micrometer-sized condensates. These opto-TopBP1 condensates are functional entities organized in tightly packed clusters of spherical nano-particles. TopBP1 condensates are reversible, occasionally fuse, and co-localize with TopBP1 partner proteins. We provide evidence that TopBP1 condensation is a molecular switch that amplifies ATR activity to phosphorylate checkpoint kinase 1 (Chk1) and slow down replication forks. Single amino acid substitutions of key residues in the intrinsically disordered ATR activation domain disrupt TopBP1 condensation and consequently ATR/Chk1 signaling. In physiologic salt concentration and pH, purified TopBP1 undergoes liquid-liquid phase separation in vitro. We propose that the actuation mechanism of ATR signaling is the assembly of TopBP1 condensates driven by highly regulated multivalent and cooperative interactions.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Carrier Proteins , Cell Nucleus , DNA-Binding Proteins , Mutation, Missense , Nuclear Proteins , Signal Transduction , Amino Acid Substitution , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Nucleus/chemistry , Cell Nucleus/genetics , Cell Nucleus/metabolism , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Sf9 Cells , Spodoptera
5.
Genomics ; 113(1 Pt 2): 707-715, 2021 01.
Article in English | MEDLINE | ID: mdl-33065246

ABSTRACT

Checkpoint kinase 1 (CHK1) is an essential kinase with a critical function in cell cycle arrest. Several potent inhibitors targeting CHK1 have been published, but most of them have failed in clinical trials. Acknowledging the emerging consequence of CHK1 inhibitors in medication of cancer, there is a demand for widening the chemical range of CHK1 inhibitors. In this research, we considered a set of in-house plant based semi-synthetic aminoarylbenzosuberene molecules as potential CHK1 inhibitors. Based on a combined computational research that consolidates molecular docking and binding free energy computations we recognized the crucial determinants for their receptor binding. The drug likeness of these molecules were also scrutinized based on their toxicity and bioavailibilty profile. The computational strategy indicates that the Bch10 could be regarded as a potential CHK1 inhibitor in comparison with top five co-crystallize molecules. Bch10 signifies a promising outlet for the development of potent inhibitors for CHK1.


Subject(s)
Antineoplastic Agents/chemistry , Checkpoint Kinase 1/antagonists & inhibitors , Coumarins/chemistry , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Humans , Protein Binding , Protein Kinase Inhibitors/pharmacology
6.
J Chem Inf Model ; 59(11): 4654-4662, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31596082

ABSTRACT

Understanding the interaction between drug molecules and proteins is one of the main challenges in drug design. Several tools have been developed recently to decrease the complexity of the process. Artificial intelligence and machine learning methods offer promising results in predicting the binding affinities. It becomes possible to do accurate predictions by using the known protein-ligand interactions. In this study, the electrostatic potential values extracted from 3-dimensional grid cubes of the drug-protein binding sites are used for predicting binding affinities of related complexes. A new algorithm with a dynamic feature selection method was implemented, which is derived from Compressed Images For Affinity Prediction (CIFAP) study, to predict binding affinities of Checkpoint Kinase 1 and Caspase 3 inhibitors.


Subject(s)
Caspase Inhibitors/pharmacology , Drug Discovery/methods , Protein Kinase Inhibitors/pharmacology , Artificial Intelligence , Binding Sites , Caspase 3/chemistry , Caspase 3/metabolism , Caspase Inhibitors/chemistry , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Drug Design , Humans , Imaging, Three-Dimensional , Ligands , Machine Learning , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemistry , Static Electricity
7.
Biosci Biotechnol Biochem ; 83(9): 1663-1675, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31094292

ABSTRACT

Members of the casein kinase 1 (CK1) family are key regulators in numerous cellular signal transduction pathways and in order to prevent the development of certain diseases, CK1 kinase activity needs to be tightly regulated. Modulation of kinase activity by site-specific phosphorylation within the C-terminal regulatory domain of CK1δ has already been shown for several cellular kinases. By using biochemical methods, we now identified residues T161, T174, T176, and S181 within the kinase domain of CK1δ as target sites for checkpoint kinase 1 (Chk1). At least residues T176 and S181 show full conservation among CK1δ orthologues from different eukaryotic species. Enzyme kinetic analysis furthermore led to the hypothesis that site-specific phosphorylation within the kinase domain finally contributes to fine-tuning of CK1δ kinase activity. These data provide a basis for the extension of our knowledge about the role of site-specific phosphorylation for regulation of CK1δ and associated signal transduction pathways.


Subject(s)
Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Humans , Phosphorylation , Signal Transduction
8.
Elife ; 82019 05 28.
Article in English | MEDLINE | ID: mdl-31135337

ABSTRACT

Coordination of the cellular response to DNA damage is organised by multi-domain 'scaffold' proteins, including 53BP1 and TOPBP1, which recognise post-translational modifications such as phosphorylation, methylation and ubiquitylation on other proteins, and are themselves carriers of such regulatory signals. Here we show that the DNA damage checkpoint regulating S-phase entry is controlled by a phosphorylation-dependent interaction of 53BP1 and TOPBP1. BRCT domains of TOPBP1 selectively bind conserved phosphorylation sites in the N-terminus of 53BP1. Mutation of these sites does not affect formation of 53BP1 or ATM foci following DNA damage, but abolishes recruitment of TOPBP1, ATR and CHK1 to 53BP1 damage foci, abrogating cell cycle arrest and permitting progression into S-phase. TOPBP1 interaction with 53BP1 is structurally complimentary to its interaction with RAD9-RAD1-HUS1, allowing these damage recognition factors to bind simultaneously to the same TOPBP1 molecule and cooperate in ATR activation in the G1 DNA damage checkpoint.


Subject(s)
Carrier Proteins/chemistry , DNA Damage/genetics , DNA-Binding Proteins/chemistry , Multiprotein Complexes/chemistry , Nuclear Proteins/chemistry , Tumor Suppressor p53-Binding Protein 1/chemistry , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Carrier Proteins/genetics , Cell Cycle Checkpoints/genetics , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Methylation , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , Phosphorylation , Protein Binding/genetics , Protein Conformation , Protein Domains/genetics , Protein Processing, Post-Translational/genetics , S Phase/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Ubiquitination/genetics
9.
Structure ; 26(8): 1137-1143.e3, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30099988

ABSTRACT

The kinase associated-1 (KA1) domain is found at the C-terminus of multiple Ser/Thr protein kinases from yeast to humans, and has been assigned autoinhibitory, membrane-binding, and substrate-targeting roles. Here, we report the crystal structure of the MARK1 kinase/UBA domain bound to its autoinhibitory KA1 domain, revealing an unexpected interface at the αD helix and contacts with both the N- and C-lobes of the kinase domain. We confirm the binding interface location in kinetic studies of variants mutated on the kinase domain surface. Together with other MARK kinase structures, the data implicate that the KA1 domain blocks peptide substrate binding. The structure highlights the kinase-specific autoinhibitory binding modes of different KA1 domains, and provides potential new avenues by which to intervene therapeutically in Alzheimer's disease and cancers in which MARK1 or related kinases are implicated.


Subject(s)
Checkpoint Kinase 1/chemistry , Peptides/chemistry , Protein Serine-Threonine Kinases/chemistry , Binding Sites , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Models, Molecular , Mutation , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein , Substrate Specificity , Thermodynamics
10.
Nature ; 557(7704): 228-232, 2018 05.
Article in English | MEDLINE | ID: mdl-29686415

ABSTRACT

Most drugs are developed through iterative rounds of chemical synthesis and biochemical testing to optimize the affinity of a particular compound for a protein target of therapeutic interest. This process is challenging because candidate molecules must be selected from a chemical space of more than 1060 drug-like possibilities 1 , and a single reaction used to synthesize each molecule has more than 107 plausible permutations of catalysts, ligands, additives and other parameters 2 . The merger of a method for high-throughput chemical synthesis with a biochemical assay would facilitate the exploration of this enormous search space and streamline the hunt for new drugs and chemical probes. Miniaturized high-throughput chemical synthesis3-7 has enabled rapid evaluation of reaction space, but so far the merger of such syntheses with bioassays has been achieved with only low-density reaction arrays, which analyse only a handful of analogues prepared under a single reaction condition8-13. High-density chemical synthesis approaches that have been coupled to bioassays, including on-bead 14 , on-surface 15 , on-DNA 16 and mass-encoding technologies 17 , greatly reduce material requirements, but they require the covalent linkage of substrates to a potentially reactive support, must be performed under high dilution and must operate in a mixture format. These reaction attributes limit the application of transition-metal catalysts, which are easily poisoned by the many functional groups present in a complex mixture, and of transformations for which the kinetics require a high concentration of reactant. Here we couple high-throughput nanomole-scale synthesis with a label-free affinity-selection mass spectrometry bioassay. Each reaction is performed at a 0.1-molar concentration in a discrete well to enable transition-metal catalysis while consuming less than 0.05 milligrams of substrate per reaction. The affinity-selection mass spectrometry bioassay is then used to rank the affinity of the reaction products to target proteins, removing the need for time-intensive reaction purification. This method enables the primary synthesis and testing steps that are critical to the invention of protein inhibitors to be performed rapidly and with minimal consumption of starting materials.


Subject(s)
Nanotechnology/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Proteins/chemistry , Biological Assay , Catalysis , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/chemistry , Drug Evaluation, Preclinical , Kinetics , Ligands , Mass Spectrometry , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Proteins/antagonists & inhibitors , Substrate Specificity
11.
J Med Chem ; 61(3): 1061-1073, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29301085

ABSTRACT

Checkpoint kinase 1 (CHK1) inhibitors are potential cancer therapeutics that can be utilized for enhancing the efficacy of DNA damaging agents. Multiple small molecule CHK1 inhibitors from different chemical scaffolds have been developed and evaluated in clinical trials in combination with chemotherapeutics and radiation treatment. Scaffold morphing of thiophene carboxamide ureas (TCUs), such as AZD7762 (1) and a related series of triazoloquinolines (TZQs), led to the identification of fused-ring bicyclic CHK1 inhibitors, 7-carboxamide thienopyridines (7-CTPs), and 7-carboxamide indoles. X-ray crystal structures reveal a key intramolecular noncovalent sulfur-oxygen interaction in aligning the hinge-binding carboxamide group to the thienopyridine core in a coplanar fashion. An intramolecular hydrogen bond to an indole NH was also effective in locking the carboxamide in the preferred bound conformation to CHK1. Optimization on the 7-CTP series resulted in the identification of lead compound 44, which displayed respectable drug-like properties and good in vitro and in vivo potency.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Drug Discovery , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1/chemistry , DNA Damage , Humans , Indoles/chemistry , Models, Molecular , Protein Domains , Pyridines/chemistry
12.
J Biol Chem ; 292(46): 19024-19033, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28972186

ABSTRACT

Precise control of the cell cycle allows for timely repair of genetic material prior to replication. One factor intimately involved in this process is checkpoint kinase 1 (Chk1), a DNA damage repair inducing Ser/Thr protein kinase that contains an N-terminal kinase domain and a C-terminal regulatory region consisting of a ∼100-residue linker followed by a putative kinase-associated 1 (KA1) domain. We report the crystal structure of the human Chk1 KA1 domain, demonstrating striking structural homology with other sequentially diverse KA1 domains. Separately purified Chk1 kinase and KA1 domains are intimately associated in solution, which results in inhibition of Chk1 kinase activity. Using truncation mutants and site-directed mutagenesis, we define the inhibitory face of the KA1 domain as a series of basic residues residing on two conserved regions of the primary structure. These findings point to KA1-mediated intramolecular autoinhibition as a key regulatory mechanism of human Chk1, and provide new therapeutic possibilities with which to attack this validated oncology target with small molecules.


Subject(s)
Checkpoint Kinase 1/chemistry , Amino Acid Sequence , Catalytic Domain , Cell Cycle , Checkpoint Kinase 1/metabolism , Crystallography, X-Ray , DNA Repair , Enzyme Activation , Humans , Models, Molecular , Protein Conformation , Sequence Alignment
13.
BMC Bioinformatics ; 18(1): 343, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28720122

ABSTRACT

BACKGROUND: In structure-based drug design, binding affinity prediction remains as a challenging goal for current scoring functions. Development of target-biased scoring functions provides a new possibility for tackling this problem, but this approach is also associated with certain technical difficulties. We previously reported the Knowledge-Guided Scoring (KGS) method as an alternative approach (BMC Bioinformatics, 2010, 11, 193-208). The key idea is to compute the binding affinity of a given protein-ligand complex based on the known binding data of an appropriate reference complex, so the error in binding affinity prediction can be reduced effectively. RESULTS: In this study, we have developed an upgraded version, i.e. KGS2, by employing 3D protein-ligand interaction fingerprints in reference selection. KGS2 was evaluated in combination with four scoring functions (X-Score, ChemPLP, ASP, and GoldScore) on five drug targets (HIV-1 protease, carbonic anhydrase 2, beta-secretase 1, beta-trypsin, and checkpoint kinase 1). In the in situ scoring test, considerable improvements were observed in most cases after application of KGS2. Besides, the performance of KGS2 was always better than KGS in all cases. In the more challenging molecular docking test, application of KGS2 also led to improved structure-activity relationship in some cases. CONCLUSIONS: KGS2 can be applied as a convenient "add-on" to current scoring functions without the need to re-engineer them, and its application is not limited to certain target proteins as customized scoring functions. As an interpolation method, its accuracy in principle can be improved further with the increasing knowledge of protein-ligand complex structures and binding affinity data. We expect that KGS2 will become a practical tool for enhancing the performance of current scoring functions in binding affinity prediction. The KGS2 software is available upon contacting the authors.


Subject(s)
Computational Biology/methods , Ligands , Proteins/chemistry , Proteins/metabolism , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Carbonic Anhydrase II/chemistry , Carbonic Anhydrase II/metabolism , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , HIV Protease/chemistry , HIV Protease/metabolism , Humans , Molecular Docking Simulation , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Protein Binding , Software
14.
J Mol Recognit ; 30(11)2017 11.
Article in English | MEDLINE | ID: mdl-28620979

ABSTRACT

Investigation of protein-ligand interactions obtained from experiments has a crucial part in the design of newly discovered and effective drugs. Analyzing the data extracted from known interactions could help scientists to predict the binding affinities of promising ligands before conducting experiments. The objective of this study is to advance the CIFAP (compressed images for affinity prediction) method, which is relevant to a protein-ligand model, identifying 2D electrostatic potential images by separating the binding site of protein-ligand complexes and using the images for predicting the computational affinity information represented by pIC50 values. The CIFAP method has 2 phases, namely, data modeling and prediction. In data modeling phase, the separated 3D structure of the binding pocket with the ligand inside is fitted into an electrostatic potential grid box, which is then compressed through 3 orthogonal directions into three 2D images for each protein-ligand complex. Sequential floating forward selection technique is performed for acquiring prediction patterns from the images. In the prediction phase, support vector regression (SVR) and partial least squares regression are used for testing the quality of the CIFAP method for predicting the binding affinity of 45 CHK1 inhibitors derived from 2-aminothiazole-4-carboxamide. The results show that the CIFAP method using both support vector regression and partial least squares regression is very effective for predicting the binding affinities of CHK1-ligand complexes with low-error values and high correlation. As a future work, the results could be improved by working on the pose of the ligands inside the grid.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Checkpoint Kinase 1/chemistry , Humans , Imaging, Three-Dimensional , Inhibitory Concentration 50 , Least-Squares Analysis , Support Vector Machine , Thiazoles/chemistry
15.
FEBS J ; 284(15): 2378-2395, 2017 08.
Article in English | MEDLINE | ID: mdl-28631426

ABSTRACT

The molecular chaperone heat shock protein 90 (Hsp90α) regulates cell proteostasis and mitigates the harmful effects of endogenous and exogenous stressors on the proteome. Indeed, the inhibition of Hsp90α ATPase activity affects the cellular response to ionizing radiation (IR). Although the interplay between Hsp90α and several DNA damage response (DDR) proteins has been reported, its role in the DDR is still unclear. Here, we show that ataxia-telangiectasia-mutated kinase (ATM) and nibrin (NBN), but not 53BP1, RAD50, and MRE11, are Hsp90α clients as the Hsp90α inhibitor 17-(allylamino)-17-demethoxygeldanamycin (17-AAG) induces ATM and NBN polyubiquitination and proteosomal degradation in normal fibroblasts and lymphoblastoid cell lines. Hsp90α-ATM and Hsp90α-NBN complexes are present in unstressed and irradiated cells, allowing the maintenance of ATM and NBN stability that is required for the MRE11/RAD50/NBN complex-dependent ATM activation and the ATM-dependent phosphorylation of both NBN and Hsp90α in response to IR-induced DNA double-strand breaks (DSBs). Hsp90α forms a complex also with ph-Ser1981-ATM following IR. Upon phosphorylation, NBN dissociates from Hsp90α and translocates at the DSBs, while phThr5/7-Hsp90α is not recruited at the damaged sites. The inhibition of Hsp90α affects nuclear localization of MRE11 and RAD50, impairs DDR signaling (e.g., BRCA1 and CHK2 phosphorylation), and slows down DSBs repair. Hsp90α inhibition does not affect DNA-dependent protein kinase (DNA-PK) activity, which possibly phosphorylates Hsp90α and H2AX after IR. Notably, Hsp90α inhibition causes H2AX phosphorylation in proliferating cells, this possibly indicating replication stress events. Overall, present data shed light on the regulatory role of Hsp90α on the DDR, controlling ATM and NBN stability and influencing the DSBs signaling and repair.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , HSP90 Heat-Shock Proteins/metabolism , Models, Biological , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Amino Acid Substitution , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Benzoquinones/pharmacology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line, Transformed , Cells, Cultured , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/chemistry , Checkpoint Kinase 2/metabolism , DNA Repair/drug effects , Gene Deletion , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Humans , Lactams, Macrocyclic/pharmacology , Nijmegen Breakage Syndrome/genetics , Nijmegen Breakage Syndrome/metabolism , Nijmegen Breakage Syndrome/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation/drug effects , Point Mutation , Proteasome Endopeptidase Complex/drug effects , Protein Multimerization/drug effects , Protein Processing, Post-Translational/drug effects , Protein Stability/drug effects , RNA Interference , Ubiquitination/drug effects
16.
BMC Cancer ; 17(1): 101, 2017 Feb 06.
Article in English | MEDLINE | ID: mdl-28166748

ABSTRACT

BACKGROUND: Body mass index (BMI) is largely investigated as a prognostic and predictive factor in triple-negative breast cancer (TNBC). Overweight and obesity are linked to a variety of pathways regulating tumor-promoting functions, including the DNA damage response (DDR). The DDR physiologically safeguards genome integrity but, in a neoplastic background, it is aberrantly engaged and protects cancer cells from chemotherapy. We herein verified the role of BMI on a previously assessed association between DDR biomarkers and pathological complete response (pCR) in TNBC patients treated with neoadjuvant chemotherapy (NACT). METHODS: In this retrospective analysis 54 TNBC patients treated with NACT were included. The relationship between DDR biomarkers, namely phosphorylated H2A Histone Family Member X (γ-H2AX) and phosphorylated checkpoint kinase 1 (pChk1), and pCR was reconsidered in light of BMI data. The Pearson's Chi-squared test of independence (2-tailed) and the Fisher Exact test were employed to assess the relationship between clinical-molecular variables and pCR. Uni- and multivariate logistic regression models were used to identify variables impacting pCR. Internal validation was carried out. RESULTS: We observed a significant association between elevated levels of the two DDR biomarkers and pCR in patients with BMI < 25 (p = 0.009 and p = 0.022 for γ-H2AX and pChk1, respectively), but not in their heavier counterpart. Results regarding γ-H2AX were confirmed in uni- and multivariate models and, again, for leaner patients only (γ-H2AXhigh vs γ-H2AXlow: OR 10.83, 95% CI: 1.79-65.55, p = 0.009). The consistency of this finding was confirmed upon internal validation. CONCLUSIONS: The predictive significance of γ-H2AX varies according to BMI status. Indeed, elevated levels of γ-H2AX seemed associated with lower pCR rate only in leaner patients, whereas differences in pCR rate according to γ-H2AX levels were not appreciable in heavier patients. Larger investigations are warranted concerning the potential role of BMI as effect modifier of the relationship between DDR-related biomarkers and clinical outcomes in TNBC.


Subject(s)
Body Mass Index , Checkpoint Kinase 1/analysis , DNA Damage , Histones/analysis , Neoadjuvant Therapy , Triple Negative Breast Neoplasms/pathology , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Female , Histones/metabolism , Humans , Middle Aged , Obesity/complications , Phosphorylation , Retrospective Studies , Triple Negative Breast Neoplasms/complications , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/therapy
17.
J Comput Aided Mol Des ; 30(12): 1149-1163, 2016 12.
Article in English | MEDLINE | ID: mdl-27722817

ABSTRACT

Ligand-based pharmacophore modeling require relatively long lists of active compounds, while a pharmacophore based on a single ligand-receptor crystallographic structure is often promiscuous. These problems prompted us to combine molecular dynamics (MD) simulation with ligand-receptor contacts analysis as means to develop valid pharmacophore model(s). The particular ligand-receptor complex is allowed to perturb over a few nano-seconds using MD simulation. Subsequently, ligand-receptor contact points (≤2.5 Å) are identified. Ligand-receptor contacts maintained above certain threshold during molecular dynamics simulation are considered critical and used to guide pharmacophore development. We termed this method as Molecular-Dynamics Based Ligand-Receptor Contact Analysis. We implemented this new methodology to develop valid pharmacophore models for check point kinase 1 (Chk1) and beta-secretase 1 (BACE1) inhibitors as case studies. The resulting pharmacophore models were validated by receiver operating characteristic curved analysis against inhibitors obtained from CHEMBL database.


Subject(s)
Amyloid Precursor Protein Secretases/chemistry , Checkpoint Kinase 1/chemistry , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Checkpoint Kinase 1/antagonists & inhibitors , Drug Design , Humans , Hydrogen Bonding , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Quantitative Structure-Activity Relationship , ROC Curve , Small Molecule Libraries/chemistry
18.
Curr Comput Aided Drug Des ; 12(4): 302-313, 2016.
Article in English | MEDLINE | ID: mdl-27585602

ABSTRACT

BACKGROUND: Checkpoint kinase 1 (Chk1) has emerged as a potential therapeutic target for design and development of novel anticancer drugs. OBJECTIVE: Herein, we have performed three-dimensional quantitative structure-activity relationship (3D-QSAR) and molecular docking analyses on a series of diazacarbazoles to design potent Chk1 inhibitors. METHODS: 3D-QSAR models were developed using comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) techniques. Docking studies were performed using AutoDock. RESULTS: The best CoMFA and CoMSIA models exhibited cross-validated correlation coefficient (q2) values of 0.631 and 0.585, and non-cross-validated correlation coefficient (r2) values of 0.933 and 0.900, respectively. CoMFA and CoMSIA models showed reasonable external predictabilities (r2 pred) of 0.672 and 0.513, respectively. CONCLUSION: A satisfactory performance in the various internal and external validation techniques indicated the reliability and robustness of the best model. Docking studies were performed to explore the binding mode of inhibitors inside the active site of Chk1. Molecular docking revealed that hydrogen bond interactions with Lys38, Glu85 and Cys87 are essential for Chk1 inhibitory activity. The binding interaction patterns observed during docking studies were complementary to 3D-QSAR results. Information obtained from the contour map analysis was utilized to design novel potent Chk1 inhibitors. Their activities and binding affinities were predicted using the derived model and docking studies. Designed inhibitors were proposed as potential candidates for experimental synthesis.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Drug Design , Molecular Docking Simulation , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Carbazoles/chemistry , Carbazoles/metabolism , Catalytic Domain , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Hydrogen Bonding , Least-Squares Analysis , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Reproducibility of Results
19.
Bioorg Med Chem Lett ; 26(17): 4310-7, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27476145

ABSTRACT

Microwave assisted synthesis of spirooxindoles via tandem double condensation between isatins and 4-hydroxycoumarin under gold catalysis is reported. The reaction is practical to perform, since the products can be isolated by simple filtration without requiring tedious column chromatography. The scope of this chemistry is exemplified by preparing structurally diverse spirooxindoles (22 examples) in excellent yields. Antimicrobial evaluation of the synthesized compounds revealed that three compounds (3a, 3f and 3s) exhibited significant MIC values in comparison to the standard drugs. Molecular docking studies of these compounds with AmpC-ß-lactamase receptor revealed that 3a exhibited minimum binding energy (-117.819kcal/mol) indicating its strong affinity towards amino acid residues via strong hydrogen bond interaction. All compounds were also evaluated for their in vitro cytotoxicity against COLO320 cancer cells. Biological assay and molecular docking studies demonstrated that 3g is the most active compound in terms of its low IC50 value (50.0µM) and least free energy of binding (-8.99kcal/mol) towards CHK1 receptor, respectively.


Subject(s)
Gold/chemistry , Indoles/chemistry , Indoles/pharmacology , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacology , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Bacteria/drug effects , Binding Sites , Catalysis , Cell Line, Tumor , Cell Survival/drug effects , Checkpoint Kinase 1/chemistry , Colonic Neoplasms/drug therapy , Coumarins/chemistry , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Microwaves , Molecular Docking Simulation , Molecular Structure , Oxindoles , Spiro Compounds/chemistry
20.
J Phys Chem B ; 120(33): 8685-95, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27258368

ABSTRACT

Mixed-solvent molecular dynamics (MixMD) is a hotspot-mapping technique that relies on molecular dynamics simulations of proteins in binary solvent mixtures. Previous work on MixMD has established the technique's effectiveness in capturing binding sites of small organic compounds. In this work, we show that MixMD can identify both competitive and allosteric sites on proteins. The MixMD approach embraces full protein flexibility and allows competition between solvent probes and water. Sites preferentially mapped by probe molecules are more likely to be binding hotspots. There are two important requirements for the identification of ligand-binding hotspots: (1) hotspots must be mapped at very high signal-to-noise ratio and (2) the hotspots must be mapped by multiple probe types. We have developed our mapping protocol around acetonitrile, isopropanol, and pyrimidine as probe solvents because they allowed us to capture hydrophilic, hydrophobic, hydrogen-bonding, and aromatic interactions. Charged probes were needed for mapping one target, and we introduce them in this work. In order to demonstrate the robust nature and wide applicability of the technique, a combined total of 5 µs of MixMD was applied across several protein targets known to exhibit allosteric modulation. Most notably, all the protein crystal structures used to initiate our simulations had no allosteric ligands bound, so there was no preorganization of the sites to predispose the simulations to find the allosteric hotspots. The protein test cases were ABL Kinase, Androgen Receptor, CHK1 Kinase, Glucokinase, PDK1 Kinase, Farnesyl Pyrophosphate Synthase, and Protein-Tyrosine Phosphatase 1B. The success of the technique is demonstrated by the fact that the top-four sites solely map the competitive and allosteric sites. Lower-ranked sites consistently map other biologically relevant sites, multimerization interfaces, or crystal-packing interfaces. Lastly, we highlight the importance of including protein flexibility by demonstrating that MixMD can map allosteric sites that are not detected in half the systems using FTMap applied to the same crystal structures.


Subject(s)
Allosteric Regulation , Catalytic Domain , Molecular Dynamics Simulation , 2-Propanol/chemistry , Acetonitriles/chemistry , Checkpoint Kinase 1/chemistry , Checkpoint Kinase 1/metabolism , Geranyltranstransferase/chemistry , Geranyltranstransferase/metabolism , Glucokinase/chemistry , Glucokinase/metabolism , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Protein Multimerization , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Pyrimidines/chemistry , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Solvents/chemistry , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...