Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000589

ABSTRACT

Mitogen-activated protein kinase kinase 1 (MAPK kinase 1, MEK1) is a key kinase in the mitogen-activated protein kinase (MAPK) signaling pathway. MEK1 mutations have been reported to lead to abnormal activation that is closely related to the malignant growth and spread of various tumors, making it an important target for cancer treatment. Targeting MEK1, four small-molecular drugs have been approved by the FDA, including Trametinib, Cobimetinib, Binimetinib, and Selumetinib. Recently, a study showed that modification with dehydroalanine (Dha) can also lead to abnormal activation of MEK1, which has the potential to promote tumor development. In this study, we used molecular dynamics simulations and metadynamics to explore the mechanism of abnormal activation of MEK1 caused by the Dha modification and predicted the inhibitory effects of four FDA-approved MEK1 inhibitors on the Dha-modified MEK1. The results showed that the mechanism of abnormal activation of MEK1 caused by the Dha modification is due to the movement of the active segment, which opens the active pocket and exposes the catalytic site, leading to sustained abnormal activation of MEK1. Among four FDA-approved inhibitors, only Selumetinib clearly blocks the active site by changing the secondary structure of the active segment from α-helix to disordered loop. Our study will help to explain the mechanism of abnormal activation of MEK1 caused by the Dha modification and provide clues for the development of corresponding inhibitors.


Subject(s)
Alanine , MAP Kinase Kinase 1 , Molecular Dynamics Simulation , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 1/chemistry , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/pharmacology , Alanine/metabolism , Humans , Catalytic Domain , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Enzyme Activation/drug effects , Benzimidazoles/pharmacology , Benzimidazoles/chemistry
2.
Nat Commun ; 15(1): 3636, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710699

ABSTRACT

Polypharmacology drugs-compounds that inhibit multiple proteins-have many applications but are difficult to design. To address this challenge we have developed POLYGON, an approach to polypharmacology based on generative reinforcement learning. POLYGON embeds chemical space and iteratively samples it to generate new molecular structures; these are rewarded by the predicted ability to inhibit each of two protein targets and by drug-likeness and ease-of-synthesis. In binding data for >100,000 compounds, POLYGON correctly recognizes polypharmacology interactions with 82.5% accuracy. We subsequently generate de-novo compounds targeting ten pairs of proteins with documented co-dependency. Docking analysis indicates that top structures bind their two targets with low free energies and similar 3D orientations to canonical single-protein inhibitors. We synthesize 32 compounds targeting MEK1 and mTOR, with most yielding >50% reduction in each protein activity and in cell viability when dosed at 1-10 µM. These results support the potential of generative modeling for polypharmacology.


Subject(s)
Molecular Docking Simulation , Humans , TOR Serine-Threonine Kinases/metabolism , Polypharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 1/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Binding , Drug Discovery/methods , Drug Design , Cell Survival/drug effects
3.
J Mol Biol ; 436(6): 168483, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38331211

ABSTRACT

RAF protein kinases are essential effectors in the MAPK pathway and are important cancer drug targets. Structural understanding of RAF activation is so far based on cryo-electron microscopy (cryo-EM) and X-ray structures of BRAF in different conformational states as inactive or active complexes with KRAS, 14-3-3 and MEK1. In this study, we have solved the first cryo-EM structures of CRAF2/14-3-32 at 3.4 Å resolution and CRAF2/14-3-32/MEK12 at 4.2 Å resolution using CRAF kinase domain expressed as constitutively active Y340D/Y341D mutant in insect cells. The overall architecture of our CRAF2/14-3-32 and CRAF2/14-3-32/MEK12 cryo-EM structures is highly similar to corresponding BRAF structures in complex with 14-3-3 or 14-3-3/MEK1 and represent the activated dimeric RAF conformation. Our CRAF cryo-EM structures provide additional insights into structural understanding of the activated CRAF2/14-3-32/MEK12 complex.


Subject(s)
14-3-3 Proteins , MAP Kinase Kinase 1 , Proto-Oncogene Proteins c-raf , Antineoplastic Agents/chemistry , Cryoelectron Microscopy , 14-3-3 Proteins/chemistry , MAP Kinase Kinase 1/chemistry , Proto-Oncogene Proteins c-raf/chemistry , Protein Conformation
4.
Cell Mol Life Sci ; 79(5): 281, 2022 May 04.
Article in English | MEDLINE | ID: mdl-35508574

ABSTRACT

MEK1 interactions with B-Raf and KSR1 are key steps in Ras/Raf/MEK/ERK signaling. Despite this, vital mechanistic details of how these execute signal transduction are still enigmatic. Among these is why, despite B-Raf and KSR1 kinase domains similarity, the B-Raf/MEK1 and KSR1/MEK1 complexes have distinct contributions to MEK1 activation, and broadly, what is KSR1's role. Our molecular dynamics simulations clarify these still unresolved ambiguities. Our results reveal that the proline-rich (P-rich) loop of MEK1 plays a decisive role in MEK1 activation loop (A-loop) phosphorylation. In the inactive B-Raf/MEK1 heterodimer, the collapsed A-loop of B-Raf interacts with the P-rich loop and A-loop of MEK1, minimizing MEK1 A-loop fluctuation and preventing it from phosphorylation. In the active B-Raf/MEK1 heterodimer, the P-rich loop moves in concert with the A-loop of B-Raf as it extends. This reduces the number of residues interacting with MEK1 A-loop, allowing increased A-loop fluctuation, and bringing Ser222 closer to ATP for phosphorylation. B-Raf αG-helix Arg662 promotes MEK1 activation by orienting Ser218 towards ATP. In KSR1/MEK1, the KSR1 αG-helix has Ala826 in place of B-Raf Arg662. This difference results in much fewer interactions between KSR1 αG-helix and MEK1 A-loop, thus a more flexible A-loop. We postulate that if KSR1 were to adopt an active configuration with an extended A-loop as seen in other protein kinases, then the MEK1 P-rich loop would extend in a similar manner, as seen in the active B-Raf/MEK1 heterodimer. This would result in highly flexible MEK1 A-loop, and KSR1 functioning as an active, B-Raf-like, kinase.


Subject(s)
Protein Kinases , Proto-Oncogene Proteins B-raf , Adenosine Triphosphate/metabolism , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , Phosphorylation , Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction
5.
Cell Biochem Funct ; 40(5): 481-490, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35604288

ABSTRACT

MEK1 is an attractive target due to its role in selective extracellular-signal-regulated kinase phosphorylation, which plays a pivotal role in regulating cell proliferation. Another benefit of targeting the MEK protein is its unique hydrophobic pocket that can accommodate highly selective allosteric inhibitors. To date, various MEK1 inhibitors have reached clinical trials against several cancers, but they were discarded due to their severe toxicity and low efficacy. Thus, the development of allosteric inhibitors for MEK1 is the demand of the hour. In this in-silico study, molecular docking, long-term molecular dynamics (5 µs), and molecular mechanics Poisson-Boltzmann surface area analysis were undertaken to address the potential of quinolines as allosteric inhibitors. We selected four reference MEK1 inhibitors for the comparative analysis. The drug-likeness and toxicity of these molecules were also examined based on their ADMET and Toxicity Prediction by Komputer Assisted Technology profiles. The outcome of the analysis revealed that the quinolines (4m, 4o, 4s, and 4n) exhibited better stability and binding affinity while being nontoxic compared to reference inhibitors. We have reached the conclusion that these quinoline molecules could be checked by experimental studies to validate their use as allosteric inhibitors against MEK1.


Subject(s)
Protein Kinase Inhibitors , Quinolines , Allosteric Site , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology
6.
Nat Commun ; 13(1): 844, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149678

ABSTRACT

The combination of ultrahigh-throughput screening and sequencing informs on function and intragenic epistasis within combinatorial protein mutant libraries. Establishing a droplet-based, in vitro compartmentalised approach for robust expression and screening of protein kinase cascades (>107 variants/day) allowed us to dissect the intrinsic molecular features of the MKK-ERK signalling pathway, without interference from endogenous cellular components. In a six-residue combinatorial library of the MKK1 docking domain, we identified 29,563 sequence permutations that allow MKK1 to efficiently phosphorylate and activate its downstream target kinase ERK2. A flexibly placed hydrophobic sequence motif emerges which is defined by higher order epistatic interactions between six residues, suggesting synergy that enables high connectivity in the sequence landscape. Through positive epistasis, MKK1 maintains function during mutagenesis, establishing the importance of co-dependent residues in mammalian protein kinase-substrate interactions, and creating a scenario for the evolution of diverse human signalling networks.


Subject(s)
Epistasis, Genetic , Mitogen-Activated Protein Kinases/chemistry , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphates/metabolism , Catalysis , Humans , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Docking Simulation , Phosphorylation , Protein Domains , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Substrate Specificity
7.
Sci Rep ; 11(1): 18366, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526571

ABSTRACT

Alterations of mitogen-activated protein kinase kinase 1 (MEK1) are commonly associated with tumorigenesis, and MEK1 is thought to be a suitable targeted therapy for various cancers. However, abnormal MEK1 alterations and their relevant clinical implications are unknown. Our research comprehensively analyzed the MEK1 alteration spectrum and provided novel insight for targeted therapies. There were 7694 samples covering 32 types of cancer from The Cancer Genome Atlas (TCGA) database. They were used to conduct an integrative analysis of MEK1 expression, alterations, functional impacts and clinical significance. There was a dramatic difference in the alteration frequency and distribution and clinical implications in 32 types of cancer from the TCGA. Skin cutaneous melanoma (SKCM) has the most alterations and has therapeutic targets located in the protein kinase domain, and the growing expression of SKCM is positively related to patient prognosis. MEK1 expression in lung adenocarcinoma (LUAD), kidney renal papillary cell carcinoma (KIRP), esophageal carcinoma (ESCA) and liver hepatocellular carcinoma (LIHC) is decreased, which is associated with better prognosis, while MEK1 expression in thymoma (THYM), stomach adenocarcinoma (STAD), kidney renal clear cell carcinoma (KIRC), testicular germ cell tumors (TGCTs) and head and neck squamous cell carcinoma (HNSC) is increased, which is associated with better prognosis. Mesothelioma (MESO) has the second highest alterations but has no therapy targets. This study provided a great and detailed interpretation of MEK1 expression, alterations and clinical implications in 32 types of cancer and reminded us to fill the gap in MEK1 research from a new perspective.


Subject(s)
MAP Kinase Kinase 1/genetics , Mutation , Neoplasms/genetics , Databases, Genetic , Humans , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , Neoplasms/pathology , Protein Domains
8.
SLAS Discov ; 26(8): 1014-1019, 2021 09.
Article in English | MEDLINE | ID: mdl-34238039

ABSTRACT

The variety and complexity of drug targets are expanding rapidly. At the same time, there is significant interest in exploring a larger chemical space to identify new candidates. Fragment-based screening (FBS) has emerged as a popular alternative to traditional high-throughput screening campaigns to identify such drug candidates. FBS identifies hit fragments that exhibit weak interactions with the target of interest, thereby enabling the rational design of small-molecule compounds from the identified hit fragments, which serve as building blocks. This strategy reduces the number of molecules to screen while also allowing the exploration of a greater chemical space.Here we use temperature-related intensity change (TRIC) technology to perform FBS against the target MAPK/ERK kinase-1 (Mek1). TRIC describes the change in fluorescence intensity of a fluorescently labeled molecule upon a change in temperature. This intensity variation is dependent on the physicochemical environment in the vicinity of the dye and strongly affected by binding events. Thus, the detection of binding events is independent of mass, making TRIC an ideal tool for FBS.Using only 150 pmol of labeled Mek1, the authors screened 193 fragments from a prescreened library in less than 1 h of measurement time, leading to 66 hits. Among those hits, they identified more than 80% of the published top hits found using orthogonal techniques. Furthermore, TRIC allowed the identification of fragments that were of poor solubility but could be mistaken as false-positive hits in other methods.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Fluorescent Dyes , Humans , Temperature
9.
J Chem Theory Comput ; 17(8): 5301-5311, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34270241

ABSTRACT

Though crucial for understanding the function of large biomolecular systems, locating the minimum free energy paths (MFEPs) between their key conformational states is far from trivial due to their high-dimensional nature. Most existing path-searching methods require a static collective variable space as input, encoding intuition or prior knowledge of the transition mechanism. Such information is, however, hardly available a priori and expensive to validate. To alleviate this issue, we have previously introduced a Traveling-salesman based Automated Path Searching method (TAPS) and demonstrated its efficiency on simple peptide systems. Having implemented a parallel version of this method, here we assess the performance of TAPS on three realistic systems (tens to hundreds of residues) in explicit solvents. We show that TAPS successfully located the MFEP for the ground/excited state transition of the T4 lysozyme L99A variant, consistent with previous findings. TAPS also helped identifying the important role of the two polar contacts in directing the loop-in/loop-out transition of the mitogen-activated protein kinase kinase (MEK1), which explained previous mutant experiments. Remarkably, at a minimal cost of 126 ns sampling, TAPS revealed that the Ltn40/Ltn10 transition of lymphotactin needs no complete unfolding/refolding of its ß-sheets and that five polar contacts are sufficient to stabilize the various partially unfolded intermediates along the MFEP. These results present TAPS as a general and promising tool for studying the functional dynamics of complex biomolecular systems.


Subject(s)
MAP Kinase Kinase 1/chemistry , Muramidase/chemistry , Lymphokines/chemistry , Lymphokines/metabolism , MAP Kinase Kinase 1/metabolism , Molecular Dynamics Simulation , Muramidase/genetics , Muramidase/metabolism , Mutagenesis, Site-Directed , Protein Conformation, beta-Strand , Protein Unfolding , Sialoglycoproteins/chemistry , Sialoglycoproteins/metabolism
10.
J Am Chem Soc ; 143(24): 9222-9229, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34121391

ABSTRACT

Development of methodologies for optically triggered protein degradation enables the study of dynamic protein functions, such as those involved in cell signaling, that are difficult to be probed with traditional genetic techniques. Here, we describe the design and implementation of a novel light-controlled peptide degron conferring N-end pathway degradation to its protein target. The degron comprises a photocaged N-terminal amino acid and a lysine-rich, 13-residue linker. By caging the N-terminal residue, we were able to optically control N-degron recognition by an E3 ligase, consequently controlling ubiquitination and proteasomal degradation of the target protein. We demonstrate broad applicability by applying this approach to a diverse set of target proteins, including EGFP, firefly luciferase, the kinase MEK1, and the phosphatase DUSP6 (also known as MKP3). The caged degron can be used with minimal protein engineering and provides virtually complete, light-triggered protein degradation on a second to minute time scale.


Subject(s)
Dual Specificity Phosphatase 6/metabolism , Green Fluorescent Proteins/metabolism , Luciferases, Firefly/metabolism , MAP Kinase Kinase 1/metabolism , Peptides/metabolism , Animals , Dual Specificity Phosphatase 6/chemistry , Fireflies , Green Fluorescent Proteins/chemistry , Humans , Luciferases, Firefly/chemistry , MAP Kinase Kinase 1/chemistry , Peptides/chemistry , Protein Conformation , Protein Engineering
11.
Biomolecules ; 11(4)2021 03 30.
Article in English | MEDLINE | ID: mdl-33808483

ABSTRACT

Mutations at different stages of the mitogen-activated protein kinase (MAPK) signaling pathway lead to aberrant activation of the involved protein kinase entities. These oncogenic modifications alter signal propagation which converge on the gatekeeper kinases MEK1/2, transmitting the input signal to ERK1/2. Thus, targeted MEK inhibition causes qualitative alterations of carcinogenic MAPK signals. Phosphorylation of the MEK1 activation loop at the positions S218 and S222 by RAF kinases triggers the conformational alignment of MEK's catalytic pocket to enable ATP-binding and substrate phosphorylation. We have extended a kinase conformation (KinCon) biosensor platform to record MEK1 activity dynamics. In addition to MEK phosphorylation by BRAF, the integration of the phosphorylation-mimetic mutations S218D/S222D triggered opening of the kinase. Structural rearrangement may involve the flexibility of the N terminal MEK1 A-helix. Application of the allosterically acting MEK inhibitors (MEKi) trametinib, cobimentinib, refametinib, and selumetinib converted activated MEK1 KinCon reporters back into a more closed inactive conformation. We confirmed MEK1 KinCon activity dynamics upon drug engagement using the patient-derived melanoma cell line A2058, which harbors the V600E hotspot BRAF mutation. In order to confirm biosensor dynamics, we simulated structure dynamics of MEK1 kinase in the presence and absence of mutations and/or MEKi binding. We observed increased dynamics for the S218D/S222D double mutant particularly in the region of the distal A-helix and alpha-C helix. These data underline that MEK1 KinCon biosensors have the potential to be subjected to MEKi efficacy validations in an intact cell setting.


Subject(s)
Drug Evaluation, Preclinical/methods , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Protein Kinase Inhibitors/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , Computer Simulation , HEK293 Cells , Humans , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , Melanoma/pathology , Molecular Dynamics Simulation , Mutation , Phosphorylation , Protein Conformation , Proto-Oncogene Proteins B-raf/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Protein Pept Lett ; 28(5): 481-488, 2021.
Article in English | MEDLINE | ID: mdl-33143608

ABSTRACT

BACKGROUND: Activation of mitogen-activated protein kinases (MAPKs) is regulated by a phosphorylation cascade comprising three kinases, MAPK kinase kinase (MAP3K), MAPK kinase (MAP2K), and MAPK. MAP2K1 and MAPK2K2, also known as MEK1 and MEK2, activate ERK1 and ERK2. The structure of the MAPK signaling cascade has been studied, but high-resolution structural studies of MAP2Ks have often focused on kinase domains or docking sites, but not on full-length proteins. OBJECTIVE: To understand the conformational dynamics of MEK1. METHODS: Full-length MEK1 was purified from Escherichia coli (BL21), and its conformational dynamics were analyzed using hydrogen/deuterium exchange mass spectrometry (HDX-MS). The effects of ATP binding were examined by co-incubating MEK1 and adenylyl-imidodiphosphate (AMP- PNP), a non-hydrolysable ATP analog. RESULTS: MEK1 exhibited mixed EX1/EX2 HDX kinetics within the N-terminal tail through ß1, αI, and the C-terminal helix. AMP-PNP binding was found to reduce conformational dynamics within the glycine-rich loop and regions near the DFG motif, along with the activation lip. CONCLUSION: We report for the first time that MEK1 has regions that slowly change its folded and unfolded states (mixed EX1/EX2 kinetics) and also report the conformational effects of ATP-binding to MEK1.


Subject(s)
Adenylyl Imidodiphosphate/chemistry , Hydrogen Deuterium Exchange-Mass Spectrometry , MAP Kinase Kinase 1/chemistry , Humans , Kinetics , Protein Domains , Protein Structure, Secondary , Recombinant Proteins
13.
Biochemistry ; 59(50): 4755-4765, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33272017

ABSTRACT

In the MAPK pathway, an oncogenic V600E mutation in B-Raf kinase causes the enzyme to be constitutively active, leading to aberrantly high phosphorylation levels of its downstream effectors, MEK and ERK kinases. The V600E mutation in B-Raf accounts for more than half of all melanomas and ∼3% of all cancers, and many drugs target the ATP binding site of the enzyme for its inhibition. Because B-Raf can develop resistance against these drugs and such drugs can induce paradoxical activation, drugs that target allosteric sites are needed. To identify other potential drug targets, we generated and kinetically characterized an active form of B-RafV600E expressed using a bacterial expression system. In doing so, we identified an α-helix on B-Raf, found at the B-Raf-MEK interface, that is critical for their interaction and the oncogenic activity of B-RafV600E. We assessed the binding between B-Raf mutants and MEK using pull downs and biolayer interferometry and assessed phosphorylation levels of MEK in vitro and in cells as well as its downstream target ERK to show that mutating certain residues on this α-helix is detrimental to binding and downstream activity. Our results suggest that this B-Raf α-helix binding site on MEK could be a site to target for drug development to treat B-RafV600E-induced melanomas.


Subject(s)
MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Allosteric Site , Amino Acid Sequence , Drug Discovery , Drug Resistance, Neoplasm , HEK293 Cells , Humans , In Vitro Techniques , Kinetics , MAP Kinase Kinase 1/genetics , MAP Kinase Signaling System , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins B-raf/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity
14.
Mol Genet Genomic Med ; 8(10): e1395, 2020 10.
Article in English | MEDLINE | ID: mdl-32757330

ABSTRACT

BACKGROUND: ß3-αC loop is a highly conserved structural domain across oncogene families, which is a switch for kinase activity. There have been numerous researches on mutations within ß3-αC loop in EGFR, but relatively less in ERBB2, BRAF, and MAP2K1. In addition, previous studies mainly focus on ß3-αC deletion in EGFR, which is the most common type affecting kinase activity and driving lung cancer. Other mutation types are not well studied. METHODS: Here we analyzed the profile of ß3-αC loop mutations in a total of 10,000 tumor biopsy and/or ctDNA patient samples using hybridization capture-based next-generation sequencing. RESULTS: We identified 1616 mutations within ß3-αC loop in this cohort. Most mutations were located in EGFR, with less percentage in ERBB2, BRAF, and MAP2K1. EGFR ß3-αC deletions occurred at a high percentage of 96.7% and were all drug-relevant. We also detected rare EGFR ß3-αC insertions and point mutations, most of which were related to EGFR TKIs resistance. ERBB2 ß3-αC deletions were only found in breast cancers and sensitive to EGFR/ERBB2 inhibitor. Moreover, BRAF and MAP2K1 mutations within ß3-αC loop also demonstrated drugs relevance. CONCLUSION: Our study showed that oncogenic mutations within the ß3-αC loop of ERBB2, MAP2K1, and BRAF are analogous to that of EGFR, which have profound effect on drug response. Understanding the mutation profile within the ß3-αC loop is critical for targeted therapies.


Subject(s)
Drug Resistance, Neoplasm , MAP Kinase Kinase 1/genetics , Mutation , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Receptor, ErbB-2/genetics , Antineoplastic Agents/therapeutic use , Conserved Sequence , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/chemistry , Neoplasms/drug therapy , Protein Domains , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/chemistry , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/chemistry
15.
Int J Mol Sci ; 21(6)2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32245216

ABSTRACT

Activation of the mitogen-activated protein kinase (MAPK) signaling pathway regulated by human MAP kinase 1 (MEK1) is associated with the carcinogenesis and progression of numerous cancers. In addition, two active mutations (P124S and E203K) have been reported to enhance the activity of MEK1, thereby eventually leading to the tumorigenesis of cancer. Trametinib is an MEK1 inhibitor for treating EML4-ALK-positive, EGFR-activated, and KRAS-mutant lung cancers. Therefore, in this study, molecular docking and molecular dynamic (MD) simulations were performed to explore the effects of inactive/active mutations (A52V/P124S and E203K) on the conformational changes of MEK1 and the changes in the interaction of MEK1 with trametinib. Moreover, steered molecular dynamic (SMD) simulations were further utilized to compare the dissociation processes of trametinib from the wild-type (WT) MEK1 and two active mutants (P124S and E203K). As a result, trametinib had stronger interactions with the non-active MEK1 (WT and A52V mutant) than the two active mutants (P124S and E203K). Moreover, two active mutants may make the allosteric channel of MEK1 wider and shorter than that of the non-active types (WT and A52V mutant). Hence, trametinib could dissociate from the active mutants (P124S and E203K) more easily compared with the WT MEK1. In summary, our theoretical results demonstrated that the active mutations may attenuate the inhibitory effects of MEK inhibitor (trametinib) on MEK1, which could be crucial clues for future anti-cancer treatment.


Subject(s)
Antineoplastic Agents/chemistry , MAP Kinase Kinase 1/chemistry , Protein Kinase Inhibitors/chemistry , Pyridones/chemistry , Pyrimidinones/chemistry , Allosteric Site/genetics , Antineoplastic Agents/pharmacology , Catalysis/drug effects , Hydrogen Bonding , Ligands , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Molecular Docking Simulation , Mutation , Protein Binding/genetics , Protein Conformation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Static Electricity , Thermodynamics
16.
Nature ; 575(7783): 545-550, 2019 11.
Article in English | MEDLINE | ID: mdl-31581174

ABSTRACT

RAF family kinases are RAS-activated switches that initiate signalling through the MAP kinase cascade to control cellular proliferation, differentiation and survival1-3. RAF activity is tightly regulated and inappropriate activation is a frequent cause of cancer4-6; however, the structural basis for RAF regulation is poorly understood at present. Here we use cryo-electron microscopy to determine autoinhibited and active-state structures of full-length BRAF in complexes with MEK1 and a 14-3-3 dimer. The reconstruction reveals an inactive BRAF-MEK1 complex restrained in a cradle formed by the 14-3-3 dimer, which binds the phosphorylated S365 and S729 sites that flank the BRAF kinase domain. The BRAF cysteine-rich domain occupies a central position that stabilizes this assembly, but the adjacent RAS-binding domain is poorly ordered and peripheral. The 14-3-3 cradle maintains autoinhibition by sequestering the membrane-binding cysteine-rich domain and blocking dimerization of the BRAF kinase domain. In the active state, these inhibitory interactions are released and a single 14-3-3 dimer rearranges to bridge the C-terminal pS729 binding sites of two BRAFs, which drives the formation of an active, back-to-back BRAF dimer. Our structural snapshots provide a foundation for understanding normal RAF regulation and its mutational disruption in cancer and developmental syndromes.


Subject(s)
14-3-3 Proteins/antagonists & inhibitors , 14-3-3 Proteins/chemistry , Cryoelectron Microscopy , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/chemistry , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Binding Sites , Cell Transformation, Neoplastic/genetics , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Domains , Protein Multimerization , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
17.
Cancer Discov ; 9(9): 1182-1191, 2019 09.
Article in English | MEDLINE | ID: mdl-31227518

ABSTRACT

We report the emergence of the novel MEK1 V211D gatekeeper mutation in a patient with BRAF K601E colon cancer treated with the allosteric MEK inhibitor binimetinib and the anti-EGFR antibody panitumumab. The MEK1 V211D mutation concurrently occurs in the same cell with BRAF K601E and leads to RAF-independent activity but remains regulated by RAF. The V211D mutation causes resistance to binimetinib by both increasing the catalytic activity of MEK1 and reducing its affinity for the drug. Moreover, the mutant exhibits reduced sensitivity to all the allosteric MEK inhibitors tested. Thus, this mutation serves as a general resistance mutation for current MEK inhibitors; however, it is sensitive to a newly reported ATP-competitive MEK inhibitor, which therefore could be used to overcome drug resistance. SIGNIFICANCE: We report a resistance mechanism to allosteric MEK inhibitors in the clinic. A MEK1 V211D mutation developed in a patient with BRAF K601E colon cancer on MEK and EGFR inhibitors. This mutant increases the catalytic activity of MEK1 and reduces its affinity for binimetinib, but remains sensitive to ATP-competitive MEK inhibitors.This article is highlighted in the In This Issue feature, p. 1143.


Subject(s)
Amino Acid Substitution , Benzimidazoles/therapeutic use , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , MAP Kinase Kinase 1/genetics , Protein Kinase Inhibitors/therapeutic use , Adult , Allosteric Regulation , Animals , Binding Sites/drug effects , Cell Line, Tumor , Colonic Neoplasms/genetics , Female , Humans , MAP Kinase Kinase 1/chemistry , Mice , NIH 3T3 Cells , Protein Binding/drug effects , Proto-Oncogene Proteins B-raf/genetics
18.
Molecules ; 24(6)2019 Mar 23.
Article in English | MEDLINE | ID: mdl-30909606

ABSTRACT

We describe herein the preparation of certain 2-substituted 3-arylquinoline derivatives and the evaluation of their anti-inflammatory effects in LPS-activated murine J774A.1 macrophage cells. Among these newly synthesized 2-substituted 3-arylquinoline derivatives, 2-(4-methoxy- benzoyl)-3-(3,4,5-trimethoxyphenyl)quinoline (18a) and 2-(4-fluorobenzoyl)-3-(3,4,5-trimethoxy- phenyl)quinoline (18b) are two of the most active compounds which can inhibit the production of NO at non-cytotoxic concentrations. Our results have also indicated that compounds 18a and 18b significantly decrease the secretion of pro-inflammatory cytokines (TNF-á and IL-6), inhibit the expression of iNOS, suppress the phosphorylation of MAPKs, and attenuate the activity of NF-êB by LPS-activated macrophages. Through molecular docking analysis, we found that 18b could fit into the middle of the TNF-á dimer and form hydrophobic interactions with Leu55, Leu57 chain A and B, Tyr59, Val123 chain B and D, Ile 155. These results suggest that both 18a and 18b are potential lead compounds in inhibiting LPS-induced inflammatory responses. Further structural optimization to discover novel anti-inflammatory agents is ongoing.


Subject(s)
Anti-Inflammatory Agents/chemistry , Inflammation/drug therapy , Macrophages/drug effects , Quinolines/chemistry , Amino Acids/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/toxicity , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/genetics , Macrophages/pathology , Mice , Molecular Docking Simulation , Nitric Oxide/metabolism , Quinolines/pharmacology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics
19.
Chembiochem ; 20(1): 66-71, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30338897

ABSTRACT

Protein phosphatase-1 (PP1)-disrupting peptides (PDPs) are selective chemical modulators of PP1 that liberate the active PP1 catalytic subunit from regulatory proteins; thus allowing the dephosphorylation of nearby substrates. We have optimized the original cell-active PDP3 for enhanced stability, and obtained insights into the chemical requirements for stabilizing this 23-mer peptide for cellular applications. The optimized PDP-Nal was used to dissect the involvement of PP1 in the MAPK signaling cascade. Specifically, we have demonstrated that, in human osteosarcoma (U2OS) cells, phosphoMEK1/2 is a direct substrate of PP1, whereas dephosphorylation of phosphoERK1/2 is indirect and likely mediated through enhanced tyrosine phosphatase activity after PDP-mediated PP1 activation. Thus, as liberators of PP1 activity, PDPs represent a valuable tool for identifying the substrates of PP1 and understanding its role in diverse signaling cascades.


Subject(s)
Peptides/metabolism , Protein Phosphatase 1/metabolism , Amino Acid Sequence , Cell Line, Tumor , Histones/chemistry , Histones/metabolism , Humans , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/chemistry , MAP Kinase Kinase Kinase 2/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation
20.
Acta Crystallogr D Struct Biol ; 74(Pt 10): 1027-1038, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30289413

ABSTRACT

The FHA domain-containing protein Mek1 is a meiosis-specific kinase that is involved in the regulation of interhomolog recombination in meiosis in Saccharomyces cerevisiae. The recruitment and activation of Mek1 require the phosphorylation of the chromosome axis protein Hop1 at Thr318 (pT318), which is necessary for recognition by the Mek1 FHA domain. Here, crystal structures of the Mek1 FHA domain in the apo state and in complex with the Hop1 pT318 peptide are presented, demonstrating that the hydrophobic residues Phe320 and Val321 at the pT+2 and pT+3 positions in the ligand contribute to the preferential recognition. It was further found that in Schizosaccharomyces pombe Mek1 FHA binds both pT15 in its N-terminal SQ/TQ cluster domain (SCD) and pT270 in the Hop1 SCD. The results revealed the structural basis for the preferential recognition of phosphorylated Hop1 by Mek1 in S. cerevisiae and facilitate the understanding of the interaction between the S. pombe Mek1 FHA domain and its binding targets.


Subject(s)
DNA-Binding Proteins/metabolism , MAP Kinase Kinase 1/metabolism , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae Proteins/metabolism , Binding Sites , DNA-Binding Proteins/chemistry , MAP Kinase Kinase 1/chemistry , Meiosis , Phosphorylation , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL