303 results on '"ras Proteins chemistry"'
Search Results
2. Positive feedback in Ras activation by full-length SOS arises from autoinhibition release mechanism.
- Author
-
Ren H, Lee AA, Lew LJN, DeGrandchamp JB, and Groves JT
- Subjects
- Kinetics, Allosteric Regulation, SOS1 Protein metabolism, SOS1 Protein chemistry, SOS1 Protein genetics, Enzyme Activation, Cell Membrane metabolism, Son of Sevenless Proteins metabolism, Son of Sevenless Proteins chemistry, Humans, Feedback, Physiological, ras Proteins metabolism, ras Proteins chemistry
- Abstract
Signaling through the Ras-MAPK pathway can exhibit switch-like activation, which has been attributed to the underlying positive feedback and bimodality in the activation of RasGDP to RasGTP by SOS. SOS contains both catalytic and allosteric Ras binding sites, and a common assumption is that allosteric activation selectively by RasGTP provides the mechanism of positive feedback. However, recent single-molecule studies have revealed that SOS catalytic rates are independent of the nucleotide state of Ras in the allosteric binding site, raising doubt about this as a positive feedback mechanism. Here, we perform detailed kinetic analyses of receptor-mediated recruitment of full-length SOS to the membrane while simultaneously monitoring its catalytic activation of Ras. These results, along with kinetic modeling, expose the autoinhibition release step in SOS, rather than either recruitment or allosteric activation, as the underlying mechanism giving rise to positive feedback in Ras activation., Competing Interests: Declaration of interests All authors declare they have no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Crystal structure of the GDP-bound human M-RAS protein in two crystal forms.
- Author
-
Bester SM, Abrahamsen R, Rodrigues Samora L, Wu WI, and Mou TC
- Subjects
- Animals, Humans, Mice, Amino Acid Sequence, Binding Sites, Crystallization, Crystallography, X-Ray, Models, Molecular, Protein Binding, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Recombinant Proteins genetics, Guanosine Diphosphate metabolism, Guanosine Diphosphate chemistry, ras Proteins chemistry, ras Proteins metabolism, ras Proteins genetics
- Abstract
M-RAS plays a crucial role in the RAF-MEK signaling pathway. When activated by GTP, M-RAS forms a complex with SHOC2 and PP1C, initiating downstream RAF-MEK signal transduction. In this study, the crystal structure of the GDP-bound human M-RAS protein is presented with two forms of crystal packing. Both the full-length and truncated human M-RAS structures aligned well with the high-confidence section of the AlphaFold2-predicted structure with low r.m.s.d., except for the Switch regions. Despite high sequence similarity to the available mouse M-RAS structure, the full-length human M-RAS structure exhibits unique crystal packing. This inactive human M-RAS structure could offer novel insights for the design of selective compounds targeting M-RAS.
- Published
- 2024
- Full Text
- View/download PDF
4. Capturing RAS oligomerization on a membrane.
- Author
-
Yun SD, Scott E, Chang JY, Bahramimoghaddam H, Lynn M, Lantz C, Russell DH, and Laganowsky A
- Subjects
- Humans, Membrane Proteins metabolism, Membrane Proteins chemistry, Membrane Proteins genetics, GTP Phosphohydrolases metabolism, GTP Phosphohydrolases chemistry, GTP Phosphohydrolases genetics, Lipoylation, ras Proteins metabolism, ras Proteins chemistry, Guanosine Triphosphate metabolism, Guanosine Diphosphate metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) chemistry, Cell Membrane metabolism, Protein Multimerization
- Abstract
RAS GTPases associate with the biological membrane where they function as molecular switches to regulate cell growth. Recent studies indicate that RAS proteins oligomerize on membranes, and disrupting these assemblies represents an alternative therapeutic strategy. However, conflicting reports on RAS assemblies, ranging in size from dimers to nanoclusters, have brought to the fore key questions regarding the stoichiometry and parameters that influence oligomerization. Here, we probe three isoforms of RAS [Kirsten Rat Sarcoma viral oncogene (KRAS), Harvey Rat Sarcoma viral oncogene (HRAS), and Neuroblastoma oncogene (NRAS)] directly from membranes using mass spectrometry. We show that KRAS on membranes in the inactive state (GDP-bound) is monomeric but forms dimers in the active state (GTP-bound). We demonstrate that the small molecule BI2852 can induce dimerization of KRAS, whereas the binding of effector proteins disrupts dimerization. We also show that RAS dimerization is dependent on lipid composition and reveal that oligomerization of NRAS is regulated by palmitoylation. By monitoring the intrinsic GTPase activity of RAS, we capture the emergence of a dimer containing either mixed nucleotides or GDP on membranes. We find that the interaction of RAS with the catalytic domain of Son of Sevenless (SOS
cat ) is influenced by membrane composition. We also capture the activation and monomer to dimer conversion of KRAS by SOScat . These results not only reveal the stoichiometry of RAS assemblies on membranes but also uncover the impact of critical factors on oligomerization, encompassing regulation by nucleotides, lipids, and palmitoylation., Competing Interests: Competing interests statement:The authors declare no competing interest.- Published
- 2024
- Full Text
- View/download PDF
5. Allosteric nanobodies to study the interactions between SOS1 and RAS.
- Author
-
Fischer B, Uchański T, Sheryazdanova A, Gonzalez S, Volkov AN, Brosens E, Zögg T, Kalichuk V, Ballet S, Versées W, Sablina AA, Pardon E, Wohlkönig A, and Steyaert J
- Subjects
- Humans, Animals, Allosteric Regulation, ras Proteins metabolism, ras Proteins chemistry, Complementarity Determining Regions chemistry, Complementarity Determining Regions immunology, Binding Sites, Camelids, New World immunology, Immunization, Signal Transduction, Models, Molecular, Single-Domain Antibodies chemistry, Single-Domain Antibodies immunology, Single-Domain Antibodies metabolism, SOS1 Protein metabolism, SOS1 Protein chemistry, SOS1 Protein genetics, SOS1 Protein immunology, Protein Binding
- Abstract
Protein-protein interactions (PPIs) are central in cell metabolism but research tools for the structural and functional characterization of these PPIs are often missing. Here we introduce broadly applicable immunization (Cross-link PPIs and immunize llamas, ChILL) and selection strategies (Display and co-selection, DisCO) for the discovery of diverse nanobodies that either stabilize or disrupt PPIs in a single experiment. We apply ChILL and DisCO to identify competitive, connective, or fully allosteric nanobodies that inhibit or facilitate the formation of the SOS1•RAS complex and modulate the nucleotide exchange rate on this pivotal GTPase in vitro as well as RAS signalling in cellulo. One of these connective nanobodies fills a cavity that was previously identified as the binding pocket for a series of therapeutic lead compounds. The long complementarity-determining region (CDR3) that penetrates this binding pocket serves as pharmacophore for extending the repertoire of potential leads., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Probing mutation-induced conformational transformation of the GTP/M-RAS complex through Gaussian accelerated molecular dynamics simulations.
- Author
-
Bao H, Wang W, Sun H, and Chen J
- Subjects
- Protein Conformation, Guanosine Triphosphate chemistry, Guanosine Triphosphate metabolism, Mutation, Molecular Dynamics Simulation, ras Proteins genetics, ras Proteins chemistry, ras Proteins metabolism
- Abstract
Mutations highly affect the structural flexibility of two switch domains in M-RAS considered an important target of anticancer drug design. Gaussian accelerated molecular dynamics (GaMD) simulations were applied to probe the effect of mutations P40D, D41E, and P40D/D41E/L51R on the conformational transition of the switch domains from the GTP-bound M-RAS. The analyses of free energy landscapes (FELs) not only reveal that three mutations induce less energetic states than the wild-type (WT) M-RAS but also verify that the switch domains are extremely disordered. Principal component analysis (PCA) and dynamics analysis suggest that three mutations greatly affect collective motions and structural flexibility of the switch domains that mostly overlap with binding regions of M-RAS to its effectors, which in turn disturbs the activity of M-RAS. The analyses of the interaction network between GTP and M-RAS show that the high instability in hydrogen bonding interactions (HBIs) of GTP with residue 41 and Y42 in the switch domain I drives the disordered states of the switch domains. This work is expected to provide a molecular mechanism for deeply understanding the function of M-RAS and future drug design towards the treatment of cancers.
- Published
- 2023
- Full Text
- View/download PDF
7. Identification of Potential Inhibitors Targeting GTPase-Kirsten RAt Sarcoma Virus (K-Ras) Driven Cancers via E-Pharmacophore-Based Virtual Screening and Drug Repurposing Approach.
- Author
-
Kumar S U, Varghese RP, Preethi VA, Doss CGP, and Zayed H
- Subjects
- Humans, Protein Binding, Pharmacophore, Clopenthixol, Drug Repositioning, Fluphenazine, Early Detection of Cancer, ras Proteins genetics, ras Proteins chemistry, Molecular Dynamics Simulation, Proto-Oncogene Proteins p21(ras) genetics, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Background: Mutations in the K-Ras gene are among the most frequent genetic alterations in various cancers, and inhibiting RAS signaling has shown promising results in treating solid tumors. However, finding effective drugs that can bind to the RAS protein remains challenging. This drove us to explore new compounds that could inhibit tumor growth, particularly in cancers that harbor K-Ras mutations., Methods: Our study used bioinformatic techniques such as E-pharmacophore virtual screening, molecular simulation, principal component analysis (PCA), extra precision (XP) docking, and ADMET analyses to identify potential inhibitors for K-Ras mutants G12C and G12D., Results: In our study, we discovered that inhibitors such as afatinib, osimertinib, and hydroxychloroquine strongly inhibit the G12C mutant. Similarly, hydroxyzine, zuclopenthixol, fluphenazine, and doxapram were potent inhibitors for the G12D mutant. Notably, all six of these molecules exhibit a high binding affinity for the H95 cryptic groove present in the mutant structure. These molecules exhibited a unique affinity mechanism at the molecular level, which was further enhanced by hydrophobic interactions. Molecular simulations and PCA revealed the formation of stable complexes within switch regions I and II. This was particularly evident in three complexes: G12C-osimertinib, G12D-fluphenazine, and G12D-zuclopenthixol. Despite the dynamic nature of switches I and II in K-Ras, the interaction of inhibitors remained stable. According to QikProp results, the properties and descriptors of the selected molecules fell within an acceptable range compared to sotorasib., Conclusions: We have successfully identified potential inhibitors of the K-Ras protein, laying the groundwork for the development of targeted therapies for cancers driven by K-Ras mutations., Competing Interests: The authors declare no conflict of interest. As C. George Priya Doss was one of guest editors of the journal, we declare that he had no involvement in the peer-review of this article and has no access to information regarding its peer-review. Full responsibility for the editorial process for this article was delegated to Milena Georgieva., (© 2023 The Author(s). Published by IMR Press.)
- Published
- 2023
- Full Text
- View/download PDF
8. Kinetic and thermodynamic allostery in the Ras protein family.
- Author
-
Manley LJ and Lin MM
- Subjects
- Kinetics, Allosteric Regulation, Molecular Dynamics Simulation, Guanosine Triphosphate metabolism, Guanosine Triphosphate chemistry, Protein Binding, Thermodynamics, ras Proteins metabolism, ras Proteins chemistry
- Abstract
Allostery, the transfer of information between distant parts of a macromolecule, is a fundamental feature of protein function and regulation. However, allosteric mechanisms are usually not explained by protein structure, requiring information on correlated fluctuations uniquely accessible to molecular simulation. Existing work to extract allosteric pathways from molecular dynamics simulations has focused on thermodynamic correlations. Here, we show how kinetic correlations encode complementary information essential to explain observed variations in allosteric regulation. We applied kinetic and thermodynamic correlation analysis on atomistic simulations of H, K, and NRas isoforms in the apo, GTP, and GDP-bound states of Ras protein, with and without complexing to its downstream effector, Raf. We show that switch I and switch II are the primary components of thermodynamic and kinetic allosteric networks, consistent with the key roles of these two motifs. These networks connect the switches to an allosteric loop recently discovered from a crystal structure of HRas. This allosteric loop is inactive in KRas, but is coupled to the hydrolysis arm switch II in NRas and HRas. We find that the mechanism in the latter two isoforms are thermodynamic and kinetic, respectively. Binding of Raf-RBD further activates thermodynamic allostery in HRas and KRas but has limited effect on NRas. These results indicate that kinetic and thermodynamic correlations are both needed to explain protein function and allostery. These two distinct channels of allosteric regulation, and their combinatorial variability, may explain how subtle mutational differences can lead to diverse regulatory profiles among enzymatic proteins., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. Structure-based prediction of Ras-effector binding affinities and design of "branchegetic" interface mutations.
- Author
-
Junk P and Kiel C
- Subjects
- Protein Binding, Mutation, Molecular Dynamics Simulation, ras Proteins genetics, ras Proteins chemistry, ras Proteins metabolism, Proteins metabolism
- Abstract
Ras is a central cellular hub protein controlling multiple cell fates. How Ras interacts with a variety of potential effector proteins is relatively unexplored, with only some key effectors characterized in great detail. Here, we have used homology modeling based on X-ray and AlphaFold2 templates to build structural models for 54 Ras-effector complexes. These models were used to estimate binding affinities using a supervised learning regressor. Furthermore, we systematically introduced Ras "branch-pruning" (or branchegetic) mutations to identify 200 interface mutations that affect the binding energy with at least one of the model structures. The impacts of these branchegetic mutants were integrated into a mathematical model to assess the potential for rewiring interactions at the Ras hub on a systems level. These findings have provided a quantitative understanding of Ras-effector interfaces and their impact on systems properties of a key cellular hub., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Unorthodox regulation of the MglA Ras-like GTPase controlling polarity in Myxococcus xanthus.
- Author
-
Dinet C and Mignot T
- Subjects
- Signal Transduction, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism, Models, Biological, Myxococcus xanthus cytology, Myxococcus xanthus enzymology, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Cell Movement, ras Proteins chemistry
- Abstract
Motile cells have developed a large array of molecular machineries to actively change their direction of movement in response to spatial cues from their environment. In this process, small GTPases act as molecular switches and work in tandem with regulators and sensors of their guanine nucleotide status (GAP, GEF, GDI and effectors) to dynamically polarize the cell and regulate its motility. In this review, we focus on Myxococcus xanthus as a model organism to elucidate the function of an atypical small Ras GTPase system in the control of directed cell motility. M. xanthus cells direct their motility by reversing their direction of movement through a mechanism involving the redirection of the motility apparatus to the opposite cell pole. The reversal frequency of moving M. xanthus cells is controlled by modular and interconnected protein networks linking the chemosensory-like frizzy (Frz) pathway - that transmits environmental signals - to the downstream Ras-like Mgl polarity control system - that comprises the Ras-like MglA GTPase protein and its regulators. Here, we discuss how variations in the GTPase interactome landscape underlie single-cell decisions and consequently, multicellular patterns., (© 2022 The Authors. FEBS Letters published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
11. Structural basis for SHOC2 modulation of RAS signalling.
- Author
-
Liau NPD, Johnson MC, Izadi S, Gerosa L, Hammel M, Bruning JM, Wendorff TJ, Phung W, Hymowitz SG, and Sudhamsu J
- Subjects
- Cryoelectron Microscopy, Guanosine Triphosphate metabolism, Humans, Multiprotein Complexes chemistry, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Multiprotein Complexes ultrastructure, Mutation, Phosphoserine, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Isoforms ultrastructure, Substrate Specificity, raf Kinases metabolism, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Protein Phosphatase 1 chemistry, Protein Phosphatase 1 genetics, Protein Phosphatase 1 metabolism, Protein Phosphatase 1 ultrastructure, Signal Transduction, ras Proteins chemistry, ras Proteins genetics, ras Proteins metabolism, ras Proteins ultrastructure
- Abstract
The RAS-RAF pathway is one of the most commonly dysregulated in human cancers
1-3 . Despite decades of study, understanding of the molecular mechanisms underlying dimerization and activation4 of the kinase RAF remains limited. Recent structures of inactive RAF monomer5 and active RAF dimer5-8 bound to 14-3-39,10 have revealed the mechanisms by which 14-3-3 stabilizes both RAF conformations via specific phosphoserine residues. Prior to RAF dimerization, the protein phosphatase 1 catalytic subunit (PP1C) must dephosphorylate the N-terminal phosphoserine (NTpS) of RAF11 to relieve inhibition by 14-3-3, although PP1C in isolation lacks intrinsic substrate selectivity. SHOC2 is as an essential scaffolding protein that engages both PP1C and RAS to dephosphorylate RAF NTpS11-13 , but the structure of SHOC2 and the architecture of the presumptive SHOC2-PP1C-RAS complex remain unknown. Here we present a cryo-electron microscopy structure of the SHOC2-PP1C-MRAS complex to an overall resolution of 3 Å, revealing a tripartite molecular architecture in which a crescent-shaped SHOC2 acts as a cradle and brings together PP1C and MRAS. Our work demonstrates the GTP dependence of multiple RAS isoforms for complex formation, delineates the RAS-isoform preference for complex assembly, and uncovers how the SHOC2 scaffold and RAS collectively drive specificity of PP1C for RAF NTpS. Our data indicate that disease-relevant mutations affect complex assembly, reveal the simultaneous requirement of two RAS molecules for RAF activation, and establish rational avenues for discovery of new classes of inhibitors to target this pathway., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
12. Structure of the MRAS-SHOC2-PP1C phosphatase complex.
- Author
-
Hauseman ZJ, Fodor M, Dhembi A, Viscomi J, Egli D, Bleu M, Katz S, Park E, Jang DM, Porter KA, Meili F, Guo H, Kerr G, Mollé S, Velez-Vega C, Beyer KS, Galli GG, Maira SM, Stams T, Clark K, Eck MJ, Tordella L, Thoma CR, and King DA
- Subjects
- 14-3-3 Proteins, Guanosine Triphosphate metabolism, Humans, MAP Kinase Signaling System, Mutation, Protein Isoforms chemistry, Protein Isoforms metabolism, Protein Subunits chemistry, Protein Subunits metabolism, raf Kinases, Crystallography, X-Ray, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins metabolism, Multiprotein Complexes chemistry, Protein Phosphatase 1 chemistry, Protein Phosphatase 1 genetics, Protein Phosphatase 1 metabolism, ras Proteins chemistry, ras Proteins metabolism
- Abstract
RAS-MAPK signalling is fundamental for cell proliferation and is altered in most human cancers
1-3 . However, our mechanistic understanding of how RAS signals through RAF is still incomplete. Although studies revealed snapshots for autoinhibited and active RAF-MEK1-14-3-3 complexes4 , the intermediate steps that lead to RAF activation remain unclear. The MRAS-SHOC2-PP1C holophosphatase dephosphorylates RAF at serine 259, resulting in the partial displacement of 14-3-3 and RAF-RAS association3,5,6 . MRAS, SHOC2 and PP1C are mutated in rasopathies-developmental syndromes caused by aberrant MAPK pathway activation6-14 -and SHOC2 itself has emerged as potential target in receptor tyrosine kinase (RTK)-RAS-driven tumours15-18 . Despite its importance, structural understanding of the SHOC2 holophosphatase is lacking. Here we determine, using X-ray crystallography, the structure of the MRAS-SHOC2-PP1C complex. SHOC2 bridges PP1C and MRAS through its concave surface and enables reciprocal interactions between all three subunits. Biophysical characterization indicates a cooperative assembly driven by the MRAS GTP-bound active state, an observation that is extendible to other RAS isoforms. Our findings support the concept of a RAS-driven and multi-molecular model for RAF activation in which individual RAS-GTP molecules recruit RAF-14-3-3 and SHOC2-PP1C to produce downstream pathway activation. Importantly, we find that rasopathy and cancer mutations reside at protein-protein interfaces within the holophosphatase, resulting in enhanced affinities and function. Collectively, our findings shed light on a fundamental mechanism of RAS biology and on mechanisms of clinically observed enhanced RAS-MAPK signalling, therefore providing the structural basis for therapeutic interventions., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
13. Structure-function analysis of the SHOC2-MRAS-PP1C holophosphatase complex.
- Author
-
Kwon JJ, Hajian B, Bian Y, Young LC, Amor AJ, Fuller JR, Fraley CV, Sykes AM, So J, Pan J, Baker L, Lee SJ, Wheeler DB, Mayhew DL, Persky NS, Yang X, Root DE, Barsotti AM, Stamford AW, Perry CK, Burgin A, McCormick F, Lemke CT, Hahn WC, and Aguirre AJ
- Subjects
- Amino Acid Motifs, Binding Sites, Guanosine Triphosphate metabolism, Humans, MAP Kinase Signaling System, Mutation, Missense, Phosphorylation, Protein Binding, Protein Stability, raf Kinases, Cryoelectron Microscopy, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism, Multiprotein Complexes ultrastructure, Protein Phosphatase 1 chemistry, Protein Phosphatase 1 metabolism, Protein Phosphatase 1 ultrastructure, ras Proteins chemistry, ras Proteins metabolism, ras Proteins ultrastructure
- Abstract
Receptor tyrosine kinase (RTK)-RAS signalling through the downstream mitogen-activated protein kinase (MAPK) cascade regulates cell proliferation and survival. The SHOC2-MRAS-PP1C holophosphatase complex functions as a key regulator of RTK-RAS signalling by removing an inhibitory phosphorylation event on the RAF family of proteins to potentiate MAPK signalling
1 . SHOC2 forms a ternary complex with MRAS and PP1C, and human germline gain-of-function mutations in this complex result in congenital RASopathy syndromes2-5 . However, the structure and assembly of this complex are poorly understood. Here we use cryo-electron microscopy to resolve the structure of the SHOC2-MRAS-PP1C complex. We define the biophysical principles of holoenzyme interactions, elucidate the assembly order of the complex, and systematically interrogate the functional consequence of nearly all of the possible missense variants of SHOC2 through deep mutational scanning. We show that SHOC2 binds PP1C and MRAS through the concave surface of the leucine-rich repeat region and further engages PP1C through the N-terminal disordered region that contains a cryptic RVXF motif. Complex formation is initially mediated by interactions between SHOC2 and PP1C and is stabilized by the binding of GTP-loaded MRAS. These observations explain how mutant versions of SHOC2 in RASopathies and cancer stabilize the interactions of complex members to enhance holophosphatase activity. Together, this integrative structure-function model comprehensively defines key binding interactions within the SHOC2-MRAS-PP1C holophosphatase complex and will inform therapeutic development ., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
14. Lipid Profiles of RAS Nanoclusters Regulate RAS Function.
- Author
-
Zhou Y and Hancock JF
- Subjects
- Animals, Cell Membrane metabolism, Humans, Models, Biological, Signal Transduction, ras Proteins chemistry, Lipids chemistry, Nanoparticles chemistry, ras Proteins metabolism
- Abstract
The lipid-anchored RAS (Rat sarcoma) small GTPases (guanosine triphosphate hydrolases) are highly prevalent in human cancer. Traditional strategies of targeting the enzymatic activities of RAS have been shown to be difficult. Alternatively, RAS function and pathology are mostly restricted to nanoclusters on the plasma membrane (PM). Lipids are important structural components of these signaling platforms on the PM. However, how RAS nanoclusters selectively enrich distinct lipids in the PM, how different lipids contribute to RAS signaling and oncogenesis and whether the selective lipid sorting of RAS nanoclusters can be targeted have not been well-understood. Latest advances in quantitative super-resolution imaging and molecular dynamic simulations have allowed detailed characterization RAS/lipid interactions. In this review, we discuss the latest findings on the select lipid composition (with headgroup and acyl chain specificities) within RAS nanoclusters, the specific mechanisms for the select lipid sorting of RAS nanoclusters on the PM and how perturbing lipid compositions within RAS nanoclusters impacts RAS function and pathology. We also describe different strategies of manipulating lipid composition within RAS nanoclusters on the PM.
- Published
- 2021
- Full Text
- View/download PDF
15. Discovery of a dual Ras and ARF6 inhibitor from a GPCR endocytosis screen.
- Author
-
Giubilaro J, Schuetz DA, Stepniewski TM, Namkung Y, Khoury E, Lara-Márquez M, Campbell S, Beautrait A, Armando S, Radresa O, Duchaine J, Lamarche-Vane N, Claing A, Selent J, Bouvier M, Marinier A, and Laporte SA
- Subjects
- ADP-Ribosylation Factor 6, ADP-Ribosylation Factors metabolism, Binding Sites, Bioluminescence Resonance Energy Transfer Techniques, Cell Line, Tumor, Cell Proliferation drug effects, Drug Discovery, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, HEK293 Cells, Humans, Molecular Dynamics Simulation, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction drug effects, ras Proteins chemistry, ras Proteins metabolism, ADP-Ribosylation Factors antagonists & inhibitors, Endocytosis drug effects, Enzyme Inhibitors pharmacology, Receptors, G-Protein-Coupled metabolism, ras Proteins antagonists & inhibitors
- Abstract
Internalization and intracellular trafficking of G protein-coupled receptors (GPCRs) play pivotal roles in cell responsiveness. Dysregulation in receptor trafficking can lead to aberrant signaling and cell behavior. Here, using an endosomal BRET-based assay in a high-throughput screen with the prototypical GPCR angiotensin II type 1 receptor (AT1R), we sought to identify receptor trafficking inhibitors from a library of ~115,000 small molecules. We identified a novel dual Ras and ARF6 inhibitor, which we named Rasarfin, that blocks agonist-mediated internalization of AT1R and other GPCRs. Rasarfin also potently inhibits agonist-induced ERK1/2 signaling by GPCRs, and MAPK and Akt signaling by EGFR, as well as prevents cancer cell proliferation. In silico modeling and in vitro studies reveal a unique binding modality of Rasarfin within the SOS-binding domain of Ras. Our findings unveil a class of dual small G protein inhibitors for receptor trafficking and signaling, useful for the inhibition of oncogenic cellular responses., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
16. A Sos proteomimetic as a pan-Ras inhibitor.
- Author
-
Hong SH, Yoo DY, Conway L, Richards-Corke KC, Parker CG, and Arora PS
- Subjects
- Animals, Biomimetics, Crystallography, X-Ray, Drug Discovery, GTP Phosphohydrolases chemistry, GTP Phosphohydrolases ultrastructure, HCT116 Cells, Helix-Loop-Helix Motifs genetics, Humans, Models, Molecular, Multiprotein Complexes chemistry, Multiprotein Complexes genetics, Proteome genetics, Signal Transduction genetics, Son of Sevenless Protein, Drosophila chemistry, Son of Sevenless Protein, Drosophila genetics, ras Proteins chemistry, ras Proteins genetics, Multiprotein Complexes ultrastructure, Protein Conformation, Son of Sevenless Protein, Drosophila ultrastructure, ras Proteins ultrastructure
- Abstract
Aberrant Ras signaling is linked to a wide spectrum of hyperproliferative diseases, and components of the signaling pathway, including Ras, have been the subject of intense and ongoing drug discovery efforts. The cellular activity of Ras is modulated by its association with the guanine nucleotide exchange factor Son of sevenless (Sos), and the high-resolution crystal structure of the Ras-Sos complex provides a basis for the rational design of orthosteric Ras ligands. We constructed a synthetic Sos protein mimic that engages the wild-type and oncogenic forms of nucleotide-bound Ras and modulates downstream kinase signaling. The Sos mimic was designed to capture the conformation of the Sos helix-loop-helix motif that makes critical contacts with Ras in its switch region. Chemoproteomic studies illustrate that the proteomimetic engages Ras and other cellular GTPases. The synthetic proteomimetic resists proteolytic degradation and enters cells through macropinocytosis. As such, it is selectively toxic to cancer cells with up-regulated macropinocytosis, including those that feature oncogenic Ras mutations., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
17. An Effective MM/GBSA Protocol for Absolute Binding Free Energy Calculations: A Case Study on SARS-CoV-2 Spike Protein and the Human ACE2 Receptor.
- Author
-
Forouzesh N and Mishra N
- Subjects
- Algorithms, Angiotensin-Converting Enzyme 2 chemistry, COVID-19 pathology, COVID-19 virology, Entropy, Humans, Ligands, Molecular Dynamics Simulation, Protein Binding, SARS-CoV-2 isolation & purification, Spike Glycoprotein, Coronavirus chemistry, raf Kinases chemistry, raf Kinases metabolism, ras Proteins chemistry, ras Proteins metabolism, Angiotensin-Converting Enzyme 2 metabolism, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus metabolism
- Abstract
The binding free energy calculation of protein-ligand complexes is necessary for research into virus-host interactions and the relevant applications in drug discovery. However, many current computational methods of such calculations are either inefficient or inaccurate in practice. Utilizing implicit solvent models in the molecular mechanics generalized Born surface area (MM/GBSA) framework allows for efficient calculations without significant loss of accuracy. Here, GBNSR6, a new flavor of the generalized Born model, is employed in the MM/GBSA framework for measuring the binding affinity between SARS-CoV-2 spike protein and the human ACE2 receptor. A computational protocol is developed based on the widely studied Ras-Raf complex, which has similar binding free energy to SARS-CoV-2/ACE2. Two options for representing the dielectric boundary of the complexes are evaluated: one based on the standard Bondi radii and the other based on a newly developed set of atomic radii (OPT1), optimized specifically for protein-ligand binding. Predictions based on the two radii sets provide upper and lower bounds on the experimental references: -14.7(ΔGbindBondi)<-10.6(ΔGbindExp.)<-4.1(ΔGbindOPT1) kcal/mol. The consensus estimates of the two bounds show quantitative agreement with the experiment values. This work also presents a novel truncation method and computational strategies for efficient entropy calculations with normal mode analysis. Interestingly, it is observed that a significant decrease in the number of snapshots does not affect the accuracy of entropy calculation, while it does lower computation time appreciably. The proposed MM/GBSA protocol can be used to study the binding mechanism of new variants of SARS-CoV-2, as well as other relevant structures.
- Published
- 2021
- Full Text
- View/download PDF
18. KRAS interaction with RAF1 RAS-binding domain and cysteine-rich domain provides insights into RAS-mediated RAF activation.
- Author
-
Tran TH, Chan AH, Young LC, Bindu L, Neale C, Messing S, Dharmaiah S, Taylor T, Denson JP, Esposito D, Nissley DV, Stephen AG, McCormick F, and Simanshu DK
- Subjects
- Binding Sites, Crystallography, X-Ray, Cysteine metabolism, Humans, Models, Molecular, Protein Binding, Protein Conformation, Protein Domains genetics, Protein Interaction Domains and Motifs, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins c-raf chemistry, Proto-Oncogene Proteins c-raf metabolism, Proto-Oncogene Proteins p21(ras) chemistry, Proto-Oncogene Proteins p21(ras) metabolism, ras Proteins chemistry, ras Proteins metabolism
- Abstract
The first step of RAF activation involves binding to active RAS, resulting in the recruitment of RAF to the plasma membrane. To understand the molecular details of RAS-RAF interaction, we present crystal structures of wild-type and oncogenic mutants of KRAS complexed with the RAS-binding domain (RBD) and the membrane-interacting cysteine-rich domain (CRD) from the N-terminal regulatory region of RAF1. Our structures reveal that RBD and CRD interact with each other to form one structural entity in which both RBD and CRD interact extensively with KRAS. Mutations at the KRAS-CRD interface result in a significant reduction in RAF1 activation despite only a modest decrease in binding affinity. Combining our structures and published data, we provide a model of RAS-RAF complexation at the membrane, and molecular insights into RAS-RAF interaction during the process of RAS-mediated RAF activation.
- Published
- 2021
- Full Text
- View/download PDF
19. The KRAS and other prenylated polybasic domain membrane anchors recognize phosphatidylserine acyl chain structure.
- Author
-
Zhou Y, Prakash PS, Liang H, Gorfe AA, and Hancock JF
- Subjects
- Animals, Cell Line, Cholesterol metabolism, Humans, Lipid Bilayers metabolism, Mutant Proteins metabolism, Nanoparticles chemistry, Static Electricity, Phosphatidylserines chemistry, Prenylation, ras Proteins chemistry, ras Proteins metabolism
- Abstract
KRAS interacts with the inner leaflet of the plasma membrane (PM) using a hybrid anchor that comprises a lysine-rich polybasic domain (PBD) and a C-terminal farnesyl chain. Electrostatic interactions have been envisaged as the primary determinant of interactions between KRAS and membranes. Here, we integrated molecular dynamics (MD) simulations and superresolution spatial analysis in mammalian cells and systematically compared four equally charged KRAS anchors: the wild-type farnesyl hexa-lysine and engineered mutants comprising farnesyl hexa-arginine, geranylgeranyl hexa-lysine, and geranylgeranyl hexa-arginine. MD simulations show that these equally charged KRAS mutant anchors exhibit distinct interactions and packing patterns with different phosphatidylserine (PtdSer) species, indicating that prenylated PBD-bilayer interactions extend beyond electrostatics. Similar observations were apparent in intact cells, where each anchor exhibited binding specificities for PtdSer species with distinct acyl chain compositions. Acyl chain composition determined responsiveness of the spatial organization of different PtdSer species to diverse PM perturbations, including transmembrane potential, cholesterol depletion, and PM curvature. In consequence, the spatial organization and PM binding of each KRAS anchor precisely reflected the behavior of its preferred PtdSer ligand to these same PM perturbations. Taken together these results show that small GTPase PBD-prenyl anchors, such as that of KRAS, have the capacity to encode binding specificity for specific acyl chains as well as lipid headgroups, which allow differential responses to biophysical perturbations that may have biological and signaling consequences for the anchored GTPase., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
20. Pan RAS-binding compounds selected from a chemical library by inhibiting interaction between RAS and a reduced affinity intracellular antibody.
- Author
-
Tanaka T, Thomas J, Van Montfort R, Miller A, and Rabbitts T
- Subjects
- Antibodies genetics, Antibodies immunology, Antibodies metabolism, Antibody Affinity, Complementarity Determining Regions chemistry, Humans, Kinetics, Mutagenesis, Site-Directed, Protein Binding, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Small Molecule Libraries chemistry, Surface Plasmon Resonance, ras Proteins chemistry, ras Proteins immunology, Small Molecule Libraries metabolism, ras Proteins metabolism
- Abstract
Intracellular antibodies are valuable tools for target validation studies for clinical situations such as cancer. Recently we have shown that antibodies can be used for drug discovery in screening for chemical compounds surrogates by showing that compounds could be developed to the so-called undruggable RAS protein family. This method, called Antibody-derived compound (Abd) technology, employed intracellular antibodies binding to RAS in a competitive surface plasmon resonance chemical library screen. Success with this method requires a high affinity interaction between the antibody and the target. We now show that reduction in the affinity (dematuration) of the anti-active RAS antibody facilitates the screening of a chemical library using an in vitro AlphaScreen method. This identified active RAS specific-binding Abd compounds that inhibit the RAS-antibody interaction. One compound is shown to be a pan-RAS binder to KRAS, HRAS and NRAS-GTP proteins with a Kd of average 37 mM, offering the possibility of a new chemical series that interacts with RAS in the switch region where the intracellular antibody binds. This simple approach shows the druggability of RAS and is generally applicable to antibody-derived chemical library screening by affording flexibility through simple antibody affinity variation. This approach can be applied to find Abd compounds as surrogates of antibody-combining sites for novel drug development in a range of human diseases.
- Published
- 2021
- Full Text
- View/download PDF
21. Interaction of Ras Binding Domain (RBD) by chemotherapeutic zinc oxide nanoparticles: Progress towards RAS pathway protein interference.
- Author
-
Mathew EN, Hurst MN, Wang B, Murthy V, Zhang Y, and DeLong RK
- Subjects
- Animals, Antineoplastic Agents chemistry, Apoptosis drug effects, Cell Line, Tumor, Mice, Protein Binding, Zinc Oxide chemistry, ras Proteins chemistry, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacology, Nanoparticles, Zinc Oxide metabolism, Zinc Oxide pharmacology, ras Proteins metabolism
- Abstract
Zinc oxide (ZnO) NP is considered as a nanoscale chemotherapeutic. Thus, the drug delivery of this inorganic NP is of considerable importance. Ras mutations are common in cancer and the activation of this signaling pathway is a hallmark in carcinoma, melanoma and many other aggressive malignancies. Thus, here we examined the binding and delivery of Ras binding domain (RBD), a model cancer-relevant protein and effector of Ras by ZnO NP. Shifts in zeta potential in water, PBS, DMEM and DMEM supplemented with FBS supported NP interaction to RBD. Fluorescence quenching of the NP was concentration-dependent for RBD, Stern-Volmer analysis of this data was used to estimate binding strength which was significant for ZnO-RBD (Kd < 10-5). ZnO NP interaction to RBD was further confirmed by pull-down assay demonstrated by SDS-PAGE analysis. The ability of ZnO NP to inhibit 3-D tumor spheroid was demonstrated in HeLa cell spheroids-the ZnO NP breaking apart these structures revealing a significant (>50%) zone of killing as shown by light and fluorescence microscopy after intra-vital staining. ZnO 100 nm was superior to ZnO 14 nm in terms of anticancer activity. When bound to ZnO NP, the anticancer activity of RBD was enhanced. These data indicate the potential diagnostic application or therapeutic activity of RBD-NP complexes in vivo which demands further investigation., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
22. Mechanisms of Ras Membrane Organization and Signaling: Ras Rocks Again.
- Author
-
Abankwa D and Gorfe AA
- Subjects
- Humans, Animals, raf Kinases metabolism, Protein Multimerization, ras Proteins metabolism, ras Proteins chemistry, Signal Transduction, Cell Membrane metabolism
- Abstract
Ras is the most frequently mutated oncogene and recent drug development efforts have spurred significant new research interest. Here we review progress toward understanding how Ras functions in nanoscale, proteo-lipid signaling complexes on the plasma membrane, called nanoclusters. We discuss how G-domain reorientation is plausibly linked to Ras-nanoclustering and -dimerization. We then look at how these mechanistic features could cooperate in the engagement and activation of RAF by Ras. Moreover, we show how this structural information can be integrated with microscopy data that provide nanoscale resolution in cell biological experiments. Synthesizing the available data, we propose to distinguish between two types of Ras nanoclusters, an active, immobile RAF-dependent type and an inactive/neutral membrane anchor-dependent. We conclude that it is possible that Ras reorientation enables dynamic Ras dimerization while the whole Ras/RAF complex transits into an active state. These transient di/oligomer interfaces of Ras may be amenable to pharmacological intervention. We close by highlighting a number of open questions including whether all effectors form active nanoclusters and whether there is an isoform specific composition of Ras nanocluster.
- Published
- 2020
- Full Text
- View/download PDF
23. A conserved, N-terminal tyrosine signal directs Ras for inhibition by Rabex-5.
- Author
-
Washington C, Chernet R, Gokhale RH, Martino-Cortez Y, Liu HY, Rosenberg AM, Shahar S, and Pfleger CM
- Subjects
- Animals, Cells, Cultured, Conserved Sequence, Drosophila, ErbB Receptors metabolism, Feedback, Physiological, Janus Kinase 2 metabolism, Phosphorylation, Tyrosine chemistry, Tyrosine genetics, Ubiquitination, ras Proteins chemistry, ras Proteins genetics, src-Family Kinases metabolism, Drosophila Proteins metabolism, Signal Transduction, Ubiquitin-Protein Ligases metabolism, ras Proteins metabolism
- Abstract
Dysregulation of the Ras oncogene in development causes developmental disorders, "Rasopathies," whereas mutational activation or amplification of Ras in differentiated tissues causes cancer. Rabex-5 (also called RabGEF1) inhibits Ras by promoting Ras mono- and di-ubiquitination. We report here that Rabex-5-mediated Ras ubiquitination requires Ras Tyrosine 4 (Y4), a site of known phosphorylation. Ras substitution mutants insensitive to Y4 phosphorylation did not undergo Rabex-5-mediated ubiquitination in cells and exhibited Ras gain-of-function phenotypes in vivo. Ras Y4 phosphomimic substitution increased Rabex-5-mediated ubiquitination in cells. Y4 phosphomimic substitution in oncogenic Ras blocked the morphological phenotypes associated with oncogenic Ras in vivo dependent on the presence of Rabex-5. We developed polyclonal antibodies raised against an N-terminal Ras peptide phosphorylated at Y4. These anti-phospho-Y4 antibodies showed dramatic recognition of recombinant wild-type Ras and RasG12V proteins when incubated with JAK2 or SRC kinases but not of RasY4F or RasY4F,G12V recombinant proteins suggesting that JAK2 and SRC could promote phosphorylation of Ras proteins at Y4 in vitro. Anti-phospho-Y4 antibodies also showed recognition of RasG12V protein, but not wild-type Ras, when incubated with EGFR. A role for JAK2, SRC, and EGFR (kinases with well-known roles to activate signaling through Ras), to promote Ras Y4 phosphorylation could represent a feedback mechanism to limit Ras activation and thus establish Ras homeostasis. Notably, rare variants of Ras at Y4 have been found in cerebellar glioblastomas. Therefore, our work identifies a physiologically relevant Ras ubiquitination signal and highlights a requirement for Y4 for Ras inhibition by Rabex-5 to maintain Ras pathway homeostasis and to prevent tissue transformation., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
24. Mechanism of adrenergic Ca V 1.2 stimulation revealed by proximity proteomics.
- Author
-
Liu G, Papa A, Katchman AN, Zakharov SI, Roybal D, Hennessey JA, Kushner J, Yang L, Chen BX, Kushnir A, Dangas K, Gygi SP, Pitt GS, Colecraft HM, Ben-Johny M, Kalocsay M, and Marx SO
- Subjects
- Animals, Calcium Channels, L-Type chemistry, Calcium Channels, N-Type metabolism, Cellular Microenvironment, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Female, HEK293 Cells, Heterotrimeric GTP-Binding Proteins metabolism, Humans, Male, Mice, Monomeric GTP-Binding Proteins metabolism, Myocardium metabolism, Phosphorylation, Protein Domains, Protein Subunits chemistry, Protein Subunits metabolism, Signal Transduction, ras Proteins chemistry, ras Proteins metabolism, Calcium Channels, L-Type metabolism, Proteomics, Receptors, Adrenergic, beta metabolism
- Abstract
Increased cardiac contractility during the fight-or-flight response is caused by β-adrenergic augmentation of Ca
V 1.2 voltage-gated calcium channels1-4 . However, this augmentation persists in transgenic murine hearts expressing mutant CaV 1.2 α1C and β subunits that can no longer be phosphorylated by protein kinase A-an essential downstream mediator of β-adrenergic signalling-suggesting that non-channel factors are also required. Here we identify the mechanism by which β-adrenergic agonists stimulate voltage-gated calcium channels. We express α1C or β2B subunits conjugated to ascorbate peroxidase5 in mouse hearts, and use multiplexed quantitative proteomics6,7 to track hundreds of proteins in the proximity of CaV 1.2. We observe that the calcium-channel inhibitor Rad8,9 , a monomeric G protein, is enriched in the CaV 1.2 microenvironment but is depleted during β-adrenergic stimulation. Phosphorylation by protein kinase A of specific serine residues on Rad decreases its affinity for β subunits and relieves constitutive inhibition of CaV 1.2, observed as an increase in channel open probability. Expression of Rad or its homologue Rem in HEK293T cells also imparts stimulation of CaV 1.3 and CaV 2.2 by protein kinase A, revealing an evolutionarily conserved mechanism that confers adrenergic modulation upon voltage-gated calcium channels.- Published
- 2020
- Full Text
- View/download PDF
25. Cysteine-based regulation of redox-sensitive Ras small GTPases.
- Author
-
Messina S, De Simone G, and Ascenzi P
- Subjects
- Animals, Cell Transformation, Neoplastic, Gene Expression Regulation, Humans, Protein Isoforms, Protein Processing, Post-Translational, Reactive Nitrogen Species metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Structure-Activity Relationship, ras Proteins chemistry, ras Proteins genetics, Cysteine metabolism, Oxidation-Reduction, ras Proteins metabolism
- Abstract
Reactive oxygen and nitrogen species (ROS and RNS, respectively) activate the redox-sensitive Ras small GTPases. The three canonical genes (HRAS, NRAS, and KRAS) are archetypes of the superfamily of small GTPases and are the most common oncogenes in human cancer. Oncogenic Ras is intimately linked to redox biology, mainly in the context of tumorigenesis. The Ras protein structure is highly conserved, especially in effector-binding regions. Ras small GTPases are redox-sensitive proteins thanks to the presence of the NKCD motif (Asn116-Lys 117-Cys118-Asp119). Notably, the ROS- and RNS-based oxidation of Cys118 affects protein stability, activity, and localization, and protein-protein interactions. Cys residues at positions 80, 181, 184, and 186 may also help modulate these actions. Moreover, oncogenic mutations of Gly12Cys and Gly13Cys may introduce additional oxidative centres and represent actionable drug targets. Here, the pathophysiological involvement of Cys-redox regulation of Ras proteins is reviewed in the context of cancer and heart and brain diseases., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. Conformational resolution of nucleotide cycling and effector interactions for multiple small GTPases determined in parallel.
- Author
-
Killoran RC and Smith MJ
- Subjects
- Guanine Nucleotide Exchange Factors chemistry, Humans, Monomeric GTP-Binding Proteins chemistry, Nucleotides chemistry, Protein Binding, Protein Conformation, Proto-Oncogene Mas, Signal Transduction, ras Proteins chemistry, rho GTP-Binding Proteins chemistry, Guanine Nucleotide Exchange Factors metabolism, Monomeric GTP-Binding Proteins metabolism, Nucleotides metabolism, ras Proteins metabolism, rho GTP-Binding Proteins metabolism
- Abstract
Small GTPases alternatively bind GDP/GTP guanine nucleotides to gate signaling pathways that direct most cellular processes. Numerous GTPases are implicated in oncogenesis, particularly the three RAS isoforms HRAS, KRAS, and NRAS and the RHO family GTPase RAC1. Signaling networks comprising small GTPases are highly connected, and there is some evidence of direct biochemical cross-talk between their functional G-domains. The activation potential of a given GTPase is contingent on a codependent interaction with the nucleotide and a Mg
2+ ion, which bind to individual variants with distinct affinities coordinated by residues in the GTPase nucleotide-binding pocket. Here, we utilized a selective-labeling strategy coupled with real-time NMR spectroscopy to monitor nucleotide exchange, GTP hydrolysis, and effector interactions of multiple small GTPases in a single complex system. We provide insight into nucleotide preference and the role of Mg2+ in activating both WT and oncogenic mutant enzymes. Multiplexing revealed guanine nucleotide exchange factor (GEF), GTPase-activating protein (GAP), and effector-binding specificities in mixtures of GTPases and resolved that the three related RAS isoforms are biochemically equivalent. This work establishes that direct quantitation of the nucleotide-bound conformation is required to accurately determine an activation potential for any given GTPase, as small GTPases such as RAS-like proto-oncogene A (RALA) or the G12C mutant of KRAS display fast exchange kinetics but have a high affinity for GDP. Furthermore, we propose that the G-domains of small GTPases behave autonomously in solution and that nucleotide cycling proceeds independently of protein concentration but is highly impacted by Mg2+ abundance., (© 2019 Killoran and Smith.)- Published
- 2019
- Full Text
- View/download PDF
27. ECOD: identification of distant homology among multidomain and transmembrane domain proteins.
- Author
-
Schaeffer RD, Kinch L, Medvedev KE, Pei J, Cheng H, and Grishin N
- Subjects
- Carrier Proteins chemistry, Cell Cycle Proteins chemistry, Crystallography, X-Ray, Databases, Protein, Endopeptidases chemistry, Escherichia coli chemistry, Humans, Leucine-tRNA Ligase chemistry, Membrane Proteins chemistry, Organic Cation Transport Proteins chemistry, Protein Multimerization, Protein Structure, Secondary, ras Proteins chemistry, ATP-Binding Cassette Transporters chemistry, ATP-Binding Cassette Transporters classification, Protein Domains, Structural Homology, Protein
- Abstract
The manual classification of protein domains is approaching its 20th anniversary. ECOD is our mixed manual-automatic domain classification. Over time, the types of proteins which require manual curation has changed. Depositions with complex multidomain and multichain arrangements are commonplace. Transmembrane domains are regularly classified. Repeatedly, domains which are initially believed to be novel are found to have homologous links to existing classified domains. Here we present a brief summary of recent manual curation efforts in ECOD generally combined with specific case studies of transmembrane and multidomain proteins wherein manual curation was useful for discovering new homologous relationships. We present a new taxonomy for the classification of ABC transporter transmembrane domains. We examine alternate topologies of the leucine-specific (LS) domain of Leucine tRNA-synthetase. Finally, we elaborate on a distant homologous links between two helical dimerization domains.
- Published
- 2019
- Full Text
- View/download PDF
28. The alteration of RhoA geranylgeranylation and Ras farnesylation breaks the integrity of the blood-testis barrier and results in hypospermatogenesis.
- Author
-
Zhu R, Wang J, Feng T, Hu X, Jiang C, Wang X, Li K, Sang Y, Hua Y, Sun H, Yao B, and Li C
- Subjects
- Animals, Azoospermia enzymology, Berberine pharmacology, Blood-Testis Barrier drug effects, Cells, Cultured, Farnesyltranstransferase genetics, Germ Cells metabolism, Humans, Male, Mice, Mice, Knockout, Mice, Transgenic, Multienzyme Complexes genetics, Sertoli Cells enzymology, Sertoli Cells metabolism, Spermatocytes metabolism, Spermatogenesis drug effects, Testis metabolism, Tight Junctions genetics, ras Proteins chemistry, ras Proteins genetics, rhoA GTP-Binding Protein chemistry, rhoA GTP-Binding Protein genetics, Azoospermia metabolism, Blood-Testis Barrier metabolism, Farnesyltranstransferase metabolism, Multienzyme Complexes metabolism, Protein Prenylation, Spermatogenesis genetics, ras Proteins metabolism, rhoA GTP-Binding Protein metabolism
- Abstract
Non-obstructive azoospermia (NOA) severely affects male infertility, however, the deep mechanisms of this disease are rarely interpreted. In this study, we find that undifferentiated spermatogonial stem cells (SSCs) still exist in the basal compartment of the seminiferous tubules and the blood-testis barrier (BTB) formed by the interaction of neighbor Sertoli cells (SCs) is incomplete in NOA patients with spermatogenic maturation arrest. The adhesions between SCs and germ cells (GCs) are also broken in NOA patients. Meanwhile, the expression level of geranylgeranyl diphosphate synthase (Ggpps), a key enzyme in mevalonate metabolic pathway, is lower in NOA patients than that in obstructive azoospermia (OA) patients. After Ggpps deletion specifically in SCs, the mice are infertile and the phenotype of the SC-Ggpps
-/- mice is similar to the NOA patients, where the BTB and the SC-GC adhesions are severely destroyed. Although SSCs are still found in the basal compartment of the seminiferous tubules, fewer mature spermatocyte and spermatid are found in SC-Ggpps-/- mice. Further examination suggests that the defect is mediated by the aberrant protein isoprenylation of RhoA and Ras family after Ggpps deletion. The exciting finding is that when the knockout mice are injected with berberine, the abnormal cell adhesions are ameliorated and spermatogenesis is partially restored. Our data suggest that the reconstruction of disrupted BTB is an effective treatment strategy for NOA patients with spermatogenic maturation arrest and hypospermatogenesis.- Published
- 2019
- Full Text
- View/download PDF
29. Predicting protein targets for drug-like compounds using transcriptomics.
- Author
-
Pabon NA, Xia Y, Estabrooks SK, Ye Z, Herbrand AK, Süß E, Biondi RM, Assimon VA, Gestwicki JE, Brodsky JL, Camacho CJ, and Bar-Joseph Z
- Subjects
- Cell Line, Computational Biology, Computer Simulation, Databases, Nucleic Acid statistics & numerical data, Drug Discovery statistics & numerical data, Drug Evaluation, Preclinical methods, Drug Evaluation, Preclinical statistics & numerical data, Gene Expression Profiling statistics & numerical data, Gene Knockdown Techniques, Gene Ontology, Gene Regulatory Networks drug effects, Humans, Models, Molecular, Molecular Docking Simulation, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacology, Proteins genetics, Ubiquitin-Protein Ligases antagonists & inhibitors, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases genetics, Wortmannin chemistry, Wortmannin pharmacology, ras Proteins antagonists & inhibitors, ras Proteins chemistry, ras Proteins genetics, Drug Discovery methods, Gene Expression Profiling methods, Proteins chemistry, Proteins drug effects
- Abstract
An expanded chemical space is essential for improved identification of small molecules for emerging therapeutic targets. However, the identification of targets for novel compounds is biased towards the synthesis of known scaffolds that bind familiar protein families, limiting the exploration of chemical space. To change this paradigm, we validated a new pipeline that identifies small molecule-protein interactions and works even for compounds lacking similarity to known drugs. Based on differential mRNA profiles in multiple cell types exposed to drugs and in which gene knockdowns (KD) were conducted, we showed that drugs induce gene regulatory networks that correlate with those produced after silencing protein-coding genes. Next, we applied supervised machine learning to exploit drug-KD signature correlations and enriched our predictions using an orthogonal structure-based screen. As a proof-of-principle for this regimen, top-10/top-100 target prediction accuracies of 26% and 41%, respectively, were achieved on a validation of set 152 FDA-approved drugs and 3104 potential targets. We then predicted targets for 1680 compounds and validated chemical interactors with four targets that have proven difficult to chemically modulate, including non-covalent inhibitors of HRAS and KRAS. Importantly, drug-target interactions manifest as gene expression correlations between drug treatment and both target gene KD and KD of genes that act up- or down-stream of the target, even for relatively weak binders. These correlations provide new insights on the cellular response of disrupting protein interactions and highlight the complex genetic phenotypes of drug treatment. With further refinement, our pipeline may accelerate the identification and development of novel chemical classes by screening compound-target interactions., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
30. De novo mutations in the GTP/GDP-binding region of RALA, a RAS-like small GTPase, cause intellectual disability and developmental delay.
- Author
-
Hiatt SM, Neu MB, Ramaker RC, Hardigan AA, Prokop JW, Hancarova M, Prchalova D, Havlovicova M, Prchal J, Stranecky V, Yim DKC, Powis Z, Keren B, Nava C, Mignot C, Rio M, Revah-Politi A, Hemati P, Stong N, Iglesias AD, Suchy SF, Willaert R, Wentzensen IM, Wheeler PG, Brick L, Kozenko M, Hurst ACE, Wheless JW, Lacassie Y, Myers RM, Barsh GS, Sedlacek Z, and Cooper GM
- Subjects
- Facies, Genotype, Guanosine Diphosphate metabolism, Guanosine Triphosphate metabolism, Humans, Mitochondrial Proteins chemistry, Models, Molecular, Mutation, Missense, Phenotype, Protein Conformation, ral GTP-Binding Proteins chemistry, ras Proteins chemistry, Developmental Disabilities genetics, Intellectual Disability genetics, Mitochondrial Proteins genetics, Mutation, Protein Interaction Domains and Motifs genetics, ral GTP-Binding Proteins genetics, ras Proteins genetics
- Abstract
Mutations that alter signaling of RAS/MAPK-family proteins give rise to a group of Mendelian diseases known as RASopathies. However, among RASopathies, the matrix of genotype-phenotype relationships is still incomplete, in part because there are many RAS-related proteins and in part because the phenotypic consequences may be variable and/or pleiotropic. Here, we describe a cohort of ten cases, drawn from six clinical sites and over 16,000 sequenced probands, with de novo protein-altering variation in RALA, a RAS-like small GTPase. All probands present with speech and motor delays, and most have intellectual disability, low weight, short stature, and facial dysmorphism. The observed rate of de novo RALA variants in affected probands is significantly higher (p = 4.93 x 10(-11)) than expected from the estimated random mutation rate. Further, all de novo variants described here affect residues within the GTP/GDP-binding region of RALA; in fact, six alleles arose at only two codons, Val25 and Lys128. The affected residues are highly conserved across both RAL- and RAS-family genes, are devoid of variation in large human population datasets, and several are homologous to positions at which disease-associated variants have been observed in other GTPase genes. We directly assayed GTP hydrolysis and RALA effector-protein binding of the observed variants, and found that all but one tested variant significantly reduced both activities compared to wild-type. The one exception, S157A, reduced GTP hydrolysis but significantly increased RALA-effector binding, an observation similar to that seen for oncogenic RAS variants. These results show the power of data sharing for the interpretation and analysis of rare variation, expand the spectrum of molecular causes of developmental disability to include RALA, and provide additional insight into the pathogenesis of human disease caused by mutations in small GTPases., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: GMC is currently serving as an Academic Editor for PLOS Genetics. GSB is currently serving as an Editor-In-Chief for PLOS Genetics. ZP is an employee of Ambry Genetics, which provides exome sequencing as a commercially available test. IMW, RW, SFS are employees of GeneDx, Inc., a wholly owned subsidiary of OPKO Health, Inc. that also offers commercial exome sequencing. The remaining authors declare no conflicts of interest.
- Published
- 2018
- Full Text
- View/download PDF
31. Potential Anti-Inflammatory and Anti-Cancer Properties of Farnesol.
- Author
-
Jung YY, Hwang ST, Sethi G, Fan L, Arfuso F, and Ahn KS
- Subjects
- Apoptosis drug effects, Cyclooxygenase 2 genetics, Farnesol chemistry, Gene Expression Regulation, Neoplastic drug effects, Humans, Inflammation pathology, Neoplasms pathology, Nitric Oxide Synthase Type II genetics, Tumor Necrosis Factor-alpha genetics, ras Proteins antagonists & inhibitors, ras Proteins chemistry, Farnesol therapeutic use, Inflammation drug therapy, Neoplasms drug therapy
- Abstract
Farnesol, an acyclic sesquiterpene alcohol, is predominantly found in essential oils of various plants in nature. It has been reported to exhibit anti-cancer and anti-inflammatory effects, and also alleviate allergic asthma, gliosis, and edema. In numerous tumor cell lines, farnesol can modulate various tumorigenic proteins and/or modulates diverse signal transduction cascades. It can also induce apoptosis and downregulate cell proliferation, angiogenesis, and cell survival. To exert its anti-inflammatory/anti-oncogenic effects, farnesol can modulate Ras protein and nuclear factor kappa-light-chain-enhancer of activated B cells activation to downregulate the expression of various inflammatory mediators such as cyclooxygenase-2, inducible nitric oxide synthase, tumor necrosis factor alpha, and interleukin-6. In this review, we describe the potential mechanisms of action underlying the therapeutic effects of farnesol against cancers and inflammatory disorders. Furthermore, these findings support the clinical development of farnesol as a potential pharmacological agent in clinical studies.
- Published
- 2018
- Full Text
- View/download PDF
32. The leucine-rich region of Flightless I interacts with R-ras to regulate cell extension formation.
- Author
-
Arora PD, He T, Ng K, and McCulloch CA
- Subjects
- Animals, Carrier Proteins chemistry, Cattle, Collagen metabolism, DNA Helicases chemistry, Mice, Microfilament Proteins, Models, Biological, Poly-ADP-Ribose Binding Proteins chemistry, Protein Binding, RNA Helicases chemistry, RNA Recognition Motif Proteins chemistry, Signal Transduction, Trans-Activators, ras Proteins chemistry, src Homology Domains, Carrier Proteins metabolism, Cell Surface Extensions metabolism, Leucine metabolism, ras Proteins metabolism
- Abstract
Flightless I (FliI) is a calcium-dependent, actin severing and capping protein that localizes to cell matrix adhesions, contributes to the generation of cell extensions, and colocalizes with Ras. Currently, the mechanism by which FliI interacts with Ras to enable assembly of actin-based cell protrusions is not defined. R-Ras, but not K-ras, H-ras, or N-ras, associated with the leucine-rich region (LRR) of FliI. Mutations of the proline-rich region of R-ras (P202A, P203A) prevented this association. Knockdown of Ras GTPase-activating SH3 domain-binding protein (G3BP1) or Rasgap
120 by small interfering RNA inhibited the formation of cell extensions and prevented interaction of R-ras and G3BP1 in FliI wild-type (WT) cells. Pull-down assays using G3BP1 fusion proteins showed a strong association of R-ras with the C-terminus of G3BP1 (amino acids 236-466), which also required the LRR of FliI. In cells that expressed the truncated N-terminus or C-terminus of G3BP1, the formation of cell extensions was blocked. Endogenous Rasgap120 interacted with the N-terminus of G3BP1 (amino acids 1-230). We conclude that in cells plated on collagen FliI-LRR interacts with R-ras to promote cell extension formation and that FliI is required for the interaction of Rasgap120 with G3BP1 to regulate R-ras activity and growth of cell extensions.- Published
- 2018
- Full Text
- View/download PDF
33. Bond swapping from a charge cloud allows flexible coordination of upstream signals through WASP: Multiple regulatory roles for the WASP basic region.
- Author
-
Tetley GJN, Szeto A, Fountain AJ, Mott HR, and Owen D
- Subjects
- Actins chemistry, Actins genetics, Amino Acid Sequence, Animals, Binding Sites, Crystallography, X-Ray, Humans, Kinetics, Neoplasms chemistry, Neoplasms pathology, Protein Binding, Regulatory Sequences, Nucleic Acid genetics, Signal Transduction, Wiskott-Aldrich Syndrome pathology, Wiskott-Aldrich Syndrome Protein genetics, cdc42 GTP-Binding Protein genetics, ras Proteins chemistry, ras Proteins genetics, Neoplasms genetics, Wiskott-Aldrich Syndrome genetics, Wiskott-Aldrich Syndrome Protein chemistry, cdc42 GTP-Binding Protein chemistry
- Abstract
Wiskott-Aldrich syndrome protein (WASP) activates the actin-related protein 2/3 homolog (Arp2/3) complex and regulates actin polymerization in a physiological setting. Cell division cycle 42 (Cdc42) is a key activator of WASP, which binds Cdc42 through a Cdc42/Rac-interactive binding (CRIB)-containing region that defines a subset of Cdc42 effectors. Here, using site-directed mutagenesis and binding affinity determination and kinetic assays, we report the results of an investigation into the energetic contributions of individual WASP residues to both the Cdc42-WASP binding interface and the kinetics of complex formation. Our results support the previously proposed dock-and-coalesce binding mechanism, initiated by electrostatic steering driven by WASP's basic region and followed by a coalescence phase likely driven by the conserved CRIB motif. The WASP basic region, however, appears also to play a role in the final complex, as its mutation affected both on- and off-rates, suggesting a more comprehensive physiological role for this region centered on the C-terminal triad of positive residues. These results highlight the expanding roles of the basic region in WASP and other CRIB-containing effector proteins in regulating complex cellular processes and coordinating multiple input signals. The data presented improve our understanding of the Cdc42-WASP interface and also add to the body of information available for Cdc42-effector complex formation, therapeutic targeting of which has promise for Ras-driven cancers. Our findings suggest that combining high-affinity peptide-binding sequences with short electrostatic steering sequences could increase the efficacy of peptidomimetic candidates designed to interfere with Cdc42 signaling in cancer., (© 2018 Tetley et al.)
- Published
- 2018
- Full Text
- View/download PDF
34. Cryo-EM structure of the active, G s -protein complexed, human CGRP receptor.
- Author
-
Liang YL, Khoshouei M, Deganutti G, Glukhova A, Koole C, Peat TS, Radjainia M, Plitzko JM, Baumeister W, Miller LJ, Hay DL, Christopoulos A, Reynolds CA, Wootten D, and Sexton PM
- Subjects
- Binding Sites, Calcitonin Gene-Related Peptide chemistry, Calcitonin Receptor-Like Protein chemistry, Calcitonin Receptor-Like Protein metabolism, GTP-Binding Protein alpha Subunits, Gs chemistry, Humans, Molecular Dynamics Simulation, Protein Domains, Protein Stability, Receptor Activity-Modifying Protein 1 chemistry, Receptor Activity-Modifying Protein 1 metabolism, Receptors, Calcitonin Gene-Related Peptide chemistry, ras Proteins chemistry, ras Proteins metabolism, Calcitonin Gene-Related Peptide metabolism, Calcitonin Receptor-Like Protein ultrastructure, Cryoelectron Microscopy, GTP-Binding Protein alpha Subunits, Gs metabolism, GTP-Binding Protein alpha Subunits, Gs ultrastructure, Receptor Activity-Modifying Protein 1 ultrastructure, Receptors, Calcitonin Gene-Related Peptide metabolism, Receptors, Calcitonin Gene-Related Peptide ultrastructure
- Abstract
Calcitonin gene-related peptide (CGRP) is a widely expressed neuropeptide that has a major role in sensory neurotransmission. The CGRP receptor is a heterodimer of the calcitonin receptor-like receptor (CLR) class B G-protein-coupled receptor and a type 1 transmembrane domain protein, receptor activity-modifying protein 1 (RAMP1). Here we report the structure of the human CGRP receptor in complex with CGRP and the G
s -protein heterotrimer at 3.3 Å global resolution, determined by Volta phase-plate cryo-electron microscopy. The receptor activity-modifying protein transmembrane domain sits at the interface between transmembrane domains 3, 4 and 5 of CLR, and stabilizes CLR extracellular loop 2. RAMP1 makes only limited direct contact with CGRP, consistent with its function in allosteric modulation of CLR. Molecular dynamics simulations indicate that RAMP1 provides stability to the receptor complex, particularly in the positioning of the extracellular domain of CLR. This work provides insights into the control of G-protein-coupled receptor function.- Published
- 2018
- Full Text
- View/download PDF
35. Small molecule inhibitors of RAS-effector protein interactions derived using an intracellular antibody fragment.
- Author
-
Quevedo CE, Cruz-Migoni A, Bery N, Miller A, Tanaka T, Petch D, Bataille CJR, Lee LYW, Fallon PS, Tulmin H, Ehebauer MT, Fernandez-Fuentes N, Russell AJ, Carr SB, Phillips SEV, and Rabbitts TH
- Subjects
- Antibodies chemistry, Biomarkers metabolism, Cell Line, Tumor, Cell Survival, Crystallography, X-Ray, HEK293 Cells, Humans, Mutation, Protein Binding, Protein Domains, Recombinant Proteins chemistry, Small Molecule Libraries, Surface Plasmon Resonance, ras Proteins chemistry, Binding Sites, Antibody, Immunoglobulin Fragments chemistry, Signal Transduction
- Abstract
Targeting specific protein-protein interactions (PPIs) is an attractive concept for drug development, but hard to implement since intracellular antibodies do not penetrate cells and most small-molecule drugs are considered unsuitable for PPI inhibition. A potential solution to these problems is to select intracellular antibody fragments to block PPIs, use these antibody fragments for target validation in disease models and finally derive small molecules overlapping the antibody-binding site. Here, we explore this strategy using an anti-mutant RAS antibody fragment as a competitor in a small-molecule library screen for identifying RAS-binding compounds. The initial hits are optimized by structure-based design, resulting in potent RAS-binding compounds that interact with RAS inside the cells, prevent RAS-effector interactions and inhibit endogenous RAS-dependent signalling. Our results may aid RAS-dependent cancer drug development and demonstrate a general concept for developing small compounds to replace intracellular antibody fragments, enabling rational drug development to target validated PPIs.
- Published
- 2018
- Full Text
- View/download PDF
36. The reactivity-driven biochemical mechanism of covalent KRAS G12C inhibitors.
- Author
-
Hansen R, Peters U, Babbar A, Chen Y, Feng J, Janes MR, Li LS, Ren P, Liu Y, and Zarrinkar PP
- Subjects
- Animals, Catalysis, Cysteine metabolism, Humans, Kinetics, Mutation, Neoplasms genetics, Protein Binding, ras Proteins chemistry, ras Proteins metabolism, Genes, ras, ras Proteins antagonists & inhibitors
- Abstract
Activating mutations in KRAS are among the most common tumor driver mutations. Until recently, KRAS had been considered undruggable with small molecules; the discovery of the covalent KRAS
G12C inhibitors ARS-853 and ARS-1620 has demonstrated that it is feasible to inhibit KRAS with high potency in cells and animals. Although the biological activity of these inhibitors has been described, the biochemical mechanism of how the compounds achieve potent inhibition remained incompletely understood. We now show that the activity of ARS-853 and ARS-1620 is primarily driven by KRAS-mediated catalysis of the chemical reaction with Cys12 in human KRASG12C , while the reversible binding affinity is weak, in the hundreds of micromolar or higher range. The mechanism resolves how an induced, shallow and dynamic pocket not expected to support high-affinity binding of small molecules can nevertheless be targeted with potent inhibitors and may be applicable to other targets conventionally considered undruggable.- Published
- 2018
- Full Text
- View/download PDF
37. Molecular recognition of RAS/RAF complex at the membrane: Role of RAF cysteine-rich domain.
- Author
-
Travers T, López CA, Van QN, Neale C, Tonelli M, Stephen AG, and Gnanakaran S
- Subjects
- Binding Sites, Cysteine metabolism, Humans, Molecular Dynamics Simulation, Protein Binding, Protein Domains, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, raf Kinases chemistry, raf Kinases genetics, ras Proteins chemistry, ras Proteins genetics, Cell Membrane metabolism, raf Kinases metabolism, ras Proteins metabolism
- Abstract
Activation of RAF kinase involves the association of its RAS-binding domain (RBD) and cysteine-rich domain (CRD) with membrane-anchored RAS. However, the overall architecture of the RAS/RBD/CRD ternary complex and the orientations of its constituent domains at the membrane remain unclear. Here, we have combined all-atom and coarse-grained molecular dynamics (MD) simulations with experimental data to construct and validate a model of membrane-anchored CRD, and used this as a basis to explore models of membrane-anchored RAS/RBD/CRD complex. First, simulations of the CRD revealed that it anchors to the membrane via insertion of its two hydrophobic loops, which is consistent with our NMR measurements of CRD bound to nanodiscs. Simulations of the CRD in the context of membrane-anchored RAS/RBD then show how CRD association with either RAS or RBD could play an unexpected role in guiding the membrane orientations of RAS/RBD. This finding has implications for the formation of RAS-RAS dimers, as different membrane orientations of RAS expose distinct putative dimerization interfaces.
- Published
- 2018
- Full Text
- View/download PDF
38. Allosteric Activation of GDP-Bound Ras Isoforms by Bisphenol Derivative Plasticisers.
- Author
-
Schöpel M, Shkura O, Seidel J, Kock K, Zhong X, Löffek S, Helfrich I, Bachmann HS, Scherkenbeck J, Herrmann C, and Stoll R
- Subjects
- Allosteric Regulation, Benzhydryl Compounds pharmacology, Endocrine Disruptors pharmacology, Guanosine Diphosphate chemistry, Guanosine Diphosphate metabolism, HeLa Cells, Humans, Phenols pharmacology, Protein Binding, ras Proteins agonists, ras Proteins metabolism, Allosteric Site, Benzhydryl Compounds chemistry, Endocrine Disruptors chemistry, Phenols chemistry, ras Proteins chemistry
- Abstract
The protein family of small GTPases controls cellular processes by acting as a binary switch between an active and an inactive state. The most prominent family members are H-Ras, N-Ras, and K-Ras isoforms, which are highly related and frequently mutated in cancer. Bisphenols are widespread in modern life because of their industrial application as plasticisers. Bisphenol A (BPA) is the best-known member and has gained significant scientific as well as public attention as an endocrine disrupting chemical, a fact that eventually led to its replacement. However, compounds used to replace BPA still contain the molecular scaffold of bisphenols. BPA, BPAF, BPB, BPE, BPF, and an amine-substituted BPAF-derivate all interact with all GDP-bound Ras-Isoforms through binding to a common site on these proteins. NMR-, SOS
cat -, and GDI- assay-based data revealed a new bisphenol-induced, allosterically activated GDP-bound Ras conformation that define these plasticisers as Ras allosteric agonists., Competing Interests: The authors declare no conflict of interest. The founding sponsors had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish the results.- Published
- 2018
- Full Text
- View/download PDF
39. The protonation states of GTP and GppNHp in Ras proteins.
- Author
-
Mann D, Güldenhaupt J, Schartner J, Gerwert K, and Kötting C
- Subjects
- Crystallography, X-Ray, Humans, Hydrogenation, Hydrolysis, Molecular Dynamics Simulation, Guanosine Triphosphate chemistry, Guanylyl Imidodiphosphate chemistry, Protons, ras Proteins chemistry
- Abstract
The small GTPase Ras transmits signals in a variety of cellular signaling pathways, most prominently in cell proliferation. GTP hydrolysis in the active center of Ras acts as a prototype for many GTPases and is the key to the understanding of several diseases, including cancer. Therefore, Ras has been the focus of intense research over the last decades. A recent neutron diffraction crystal structure of Ras indicated a protonated γ-guanylyl imidodiphosphate (γ-GppNHp) group, which has put the protonation state of GTP in question. A possible protonation of GTP was not considered in previously published mechanistic studies. To determine the detailed prehydrolysis state of Ras, we calculated infrared and NMR spectra from quantum mechanics/molecular mechanics (QM/MM) simulations and compared them with those from previous studies. Furthermore, we measured infrared spectra of GTP and several GTP analogs bound to lipidated Ras on a membrane system under near-native conditions. Our findings unify results from previous studies and indicate a structural model confirming the hypothesis that γ-GTP is fully deprotonated in the prehydrolysis state of Ras., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
40. Exceptionally high-affinity Ras binders that remodel its effector domain.
- Author
-
McGee JH, Shim SY, Lee SJ, Swanson PK, Jiang SY, Durney MA, and Verdine GL
- Subjects
- Amino Acid Sequence, Databases, Protein, Drug Discovery, Models, Molecular, Mutation, Protein Binding, Protein Domains drug effects, Protein Multimerization, Protein Structure, Quaternary, Proteins chemistry, Proteins genetics, Proteins metabolism, Proteins pharmacology, ras Proteins chemistry, ras Proteins metabolism
- Abstract
The Ras proteins are aberrantly activated in a wide range of human cancers, often endowing tumors with aggressive properties and resistance to therapy. Decades of effort to develop direct Ras inhibitors for clinical use have thus far failed, largely because of a lack of adequate small-molecule-binding pockets on the Ras surface. Here, we report the discovery of Ras-binding miniproteins from a naïve library and their evolution to afford versions with midpicomolar affinity to Ras. A series of biochemical experiments indicated that these miniproteins bind to the Ras effector domain as dimers, and high-resolution crystal structures revealed that these miniprotein dimers bind Ras in an unprecedented mode in which the Ras effector domain is remodeled to expose an extended pocket that connects two isolated pockets previously found to engage small-molecule ligands. We also report a Ras point mutant that stabilizes the protein in the open conformation trapped by these miniproteins. These findings provide new tools for studying Ras structure and function and present opportunities for the development of both miniprotein and small-molecule inhibitors that directly target the Ras proteins., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
41. Deciphering lipid codes: K-Ras as a paradigm.
- Author
-
Zhou Y and Hancock JF
- Subjects
- Animals, Cell Membrane metabolism, Humans, MAP Kinase Signaling System, ras Proteins chemistry, ras Proteins genetics, Membrane Lipids metabolism, ras Proteins metabolism
- Abstract
The cell plasma membrane (PM) is a highly dynamic and heterogeneous lipid environment, driven by complex hydrophobic and electrostatic interactions among the hundreds of types of lipid species. Although the biophysical processes governing lipid lateral segregation in the cell PM have been established in vitro, biological implications of lipid heterogeneity are poorly understood. Of particular interest is how membrane proteins potentially utilize transient spatial clustering of PM lipids to regulate function. This review focuses on a lipid-anchored small GTPase K-Ras as an example to explore how its C-terminal membrane-anchoring domain, consisting of a contiguous hexa-lysine polybasic domain and an adjacent farnesyl anchor, possesses a complex coding mechanism for highly selective lipid sorting on the PM. How this lipid specificity modulates K-Ras signal transmission will also be discussed., (© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
42. Large scale analysis of protein conformational transitions from aqueous to non-aqueous media.
- Author
-
Rueda AJV, Monzon AM, Ardanaz SM, Iglesias LE, and Parisi G
- Subjects
- Biocatalysis, Databases, Protein, Humans, Protein Structure, Secondary, Protein Structure, Tertiary, Proteins metabolism, ras Proteins chemistry, ras Proteins metabolism, Computational Biology methods, Proteins chemistry, Solvents chemistry, Water chemistry
- Abstract
Background: Biocatalysis in organic solvents is nowadays a common practice with a large potential in Biotechnology. Several studies report that proteins which are co-crystallized or soaked in organic solvents preserve their fold integrity showing almost identical arrangements when compared to their aqueous forms. However, it is well established that the catalytic activity of proteins in organic solvents is much lower than in water. In order to explain this diminished activity and to further characterize the behaviour of proteins in non-aqueous environments, we performed a large-scale analysis (1737 proteins) of the conformational diversity of proteins crystallized in aqueous and co-crystallized or soaked in non-aqueous media., Results: Using proteins' experimentally determined conformational diversity taken from CoDNaS database, we found that proteins in non-aqueous media display much lower conformational diversity when compared to the corresponding conformers obtained in water. When conformational diversity is compared between conformers obtained in different non-aqueous media, their structural differences are larger and mostly independent of the presence of cognate ligands. We also found that conformers corresponding to non-aqueous media have larger but less flexible cavities, lower number of disordered regions and lower active-site residue mobility., Conclusions: Our results show that non-aqueous media conformers have specific structural features and that they do not adopt extreme conformations found in aqueous media. This makes them clearly different from their corresponding aqueous conformers.
- Published
- 2018
- Full Text
- View/download PDF
43. The 150 most important questions in cancer research and clinical oncology series: questions 67-75 : Edited by Chinese Journal of Cancer.
- Subjects
- Animals, Biomedical Research, Chromosomal Instability, Drug Resistance, Neoplasm, Early Detection of Cancer, Heavy Ion Radiotherapy, Humans, Medical Oncology, Mutation, Neoplastic Stem Cells, ras Proteins chemistry, ras Proteins metabolism, Neoplasms diagnosis, Neoplasms genetics, Neoplasms metabolism, Neoplasms therapy
- Abstract
Since the beginning of 2017, Chinese Journal of Cancer has published a series of important questions in cancer research and clinical oncology, which sparkle diverse thoughts, interesting communications, and potential collaborations among researchers all over the world. In this article, 9 more questions are presented as followed. Question 67. How could we overcome the resistance of hepatocellular carcinoma against chemotherapeutics? Question 68. Is pursuit of non-covalent small-molecule binders of RAS proteins viable as a strategy of cancer drug discovery? Question 69. In what oligomeric structures do RAS proteins signal? Question 70. How can we achieve non-invasive early detection and diagnosis of lung cancer? Question 71. Does genetic information influence the volatolome enabling diagnosis of lung cancer with genetic mutations via cell headspace or breath analysis? Question 72. Is heavy ion beam radiotherapy effective to kill cancer stem cells? Question 73. Is there any diversity among different types of cancer in terms of sensitivity to heavy ion beam radiotherapy? Question 74. Can targeted alpha-particle therapy augment the effect of carbon ion radiotherapy on malignancies? Question 75. How does chromosomal instability drive tumor progression?
- Published
- 2017
- Full Text
- View/download PDF
44. Molecular Dynamics Simulations and Dynamic Network Analysis Reveal the Allosteric Unbinding of Monobody to H-Ras Triggered by R135K Mutation.
- Author
-
Ni D, Song K, Zhang J, and Lu S
- Subjects
- Algorithms, Allosteric Regulation, Humans, Models, Molecular, Protein Binding, Protein Interaction Domains and Motifs, Structure-Activity Relationship, Amino Acid Substitution, Molecular Dynamics Simulation, Mutation, Protein Conformation, ras Proteins chemistry, ras Proteins genetics
- Abstract
Ras proteins, as small GTPases, mediate cell proliferation, survival and differentiation. Ras mutations have been associated with a broad spectrum of human cancers and thus targeting Ras represents a potential way forward for cancer therapy. A recently reported monobody NS1 allosterically disrupts the Ras-mediated signaling pathway, but its efficacy is reduced by R135K mutation in H-Ras. However, the detailed mechanism is unresolved. Here, using molecular dynamics (MD) simulations and dynamic network analysis, we explored the molecular mechanism for the unbinding of NS1 to H-Ras and shed light on the underlying allosteric network in H-Ras. MD simulations revealed that the overall structures of the two complexes did not change significantly, but the H-Ras-NS1 interface underwent significant conformational alteration in the mutant Binding free energy analysis showed that NS1 binding was unfavored after R135K mutation, which resulted in the unfavorable binding of NS1. Furthermore, the critical residues on H-Ras responsible for the loss of binding of NS1 were identified. Importantly, the allosteric networks for these important residues were revealed, which yielded a novel insight into the allosteric regulatory mechanism of H-Ras., Competing Interests: The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
45. Intrinsic protein disorder in oncogenic KRAS signaling.
- Author
-
Nussinov R, Jang H, Tsai CJ, Liao TJ, Li S, Fushman D, and Zhang J
- Subjects
- Calmodulin chemistry, Calmodulin metabolism, Intrinsically Disordered Proteins chemistry, Lipoylation, Molecular Dynamics Simulation, Neoplasms pathology, Protein Domains, Protein Isoforms chemistry, Protein Isoforms metabolism, Signal Transduction, raf Kinases chemistry, raf Kinases metabolism, ras Proteins chemistry, Intrinsically Disordered Proteins metabolism, Neoplasms metabolism, ras Proteins metabolism
- Abstract
How Ras, and in particular its most abundant oncogenic isoform K-Ras4B, is activated and signals in proliferating cells, poses some of the most challenging questions in cancer cell biology. In this paper, we ask how intrinsically disordered regions in K-Ras4B and its effectors help promote proliferative signaling. Conformational disorder allows spanning long distances, supports hinge motions, promotes anchoring in membranes, permits segments to fulfil multiple roles, and broadly is crucial for activation mechanisms and intensified oncogenic signaling. Here, we provide an overview illustrating some of the key mechanisms through which conformational disorder can promote oncogenesis, with K-Ras4B signaling serving as an example. We discuss (1) GTP-bound KRas4B activation through membrane attachment; (2) how farnesylation and palmitoylation can promote isoform functional specificity; (3) calmodulin binding and PI3K activation; (4) how Ras activates its RASSF5 cofactor, thereby stimulating signaling of the Hippo pathway and repressing proliferation; and (5) how intrinsically disordered segments in Raf help its attachment to the membrane and activation. Collectively, we provide the first inclusive review of the roles of intrinsic protein disorder in oncogenic Ras-driven signaling. We believe that a broad picture helps to grasp and formulate key mechanisms in Ras cancer biology and assists in therapeutic intervention.
- Published
- 2017
- Full Text
- View/download PDF
46. Structural basis for intramolecular interaction of post-translationally modified H-Ras•GTP prepared by protein ligation.
- Author
-
Ke H, Matsumoto S, Murashima Y, Taniguchi-Tamura H, Miyamoto R, Yoshikawa Y, Tsuda C, Kumasaka T, Mizohata E, Edamatsu H, and Kataoka T
- Subjects
- Amino Acid Sequence, Catalytic Domain, Crystallography, X-Ray, HEK293 Cells, Humans, Models, Molecular, Protein Binding, Guanosine Triphosphate metabolism, Protein Processing, Post-Translational, ras Proteins chemistry, ras Proteins metabolism
- Abstract
Ras undergoes post-translational modifications including farnesylation, proteolysis, and carboxymethylation at the C terminus, which are necessary for membrane recruitment and effector recognition. Full activation of c-Raf-1 requires cooperative interaction of the farnesylated C terminus and the activator region of Ras with its cysteine-rich domain (CRD). However, the molecular basis for this interaction remains unclear because of difficulties in preparing modified Ras in amounts sufficient for structural studies. Here, we use Sortase A-catalyzed protein ligation to prepare modified Ras in sufficient amounts for NMR and X-ray crystallographic analyses. The results show that the farnesylated C terminus establishes an intramolecular interaction with the catalytic domain and brings the farnesyl moiety to the proximity of the activator region, which may be responsible for their cooperative recognition of c-Raf-1-CRD., (© 2017 Federation of European Biochemical Societies.)
- Published
- 2017
- Full Text
- View/download PDF
47. RAS Proteins and Their Regulators in Human Disease.
- Author
-
Simanshu DK, Nissley DV, and McCormick F
- Subjects
- Animals, Cell Membrane metabolism, Congenital Abnormalities metabolism, Humans, Mental Disorders metabolism, Mutation, Neoplasms metabolism, Phylogeny, Signal Transduction, Yeasts, ras Proteins chemistry, ras Proteins genetics, ras Proteins metabolism
- Abstract
RAS proteins are binary switches, cycling between ON and OFF states during signal transduction. These switches are normally tightly controlled, but in RAS-related diseases, such as cancer, RASopathies, and many psychiatric disorders, mutations in the RAS genes or their regulators render RAS proteins persistently active. The structural basis of the switch and many of the pathways that RAS controls are well known, but the precise mechanisms by which RAS proteins function are less clear. All RAS biology occurs in membranes: a precise understanding of RAS' interaction with membranes is essential to understand RAS action and to intervene in RAS-driven diseases., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Mechanism of SOS PR-domain autoinhibition revealed by single-molecule assays on native protein from lysate.
- Author
-
Lee YK, Low-Nam ST, Chung JK, Hansen SD, Lam HYM, Alvarez S, and Groves JT
- Subjects
- Allosteric Site, Binding Sites, Cell Membrane metabolism, GRB2 Adaptor Protein chemistry, GRB2 Adaptor Protein metabolism, HEK293 Cells, Humans, Microscopy, Fluorescence, Protein Binding, ras Proteins chemistry, ras Proteins metabolism, Models, Molecular, Protein Domains, SOS1 Protein chemistry, SOS1 Protein metabolism
- Abstract
The guanine nucleotide exchange factor (GEF) Son of Sevenless (SOS) plays a critical role in signal transduction by activating Ras. Here we introduce a single-molecule assay in which individual SOS molecules are captured from raw cell lysate using Ras-functionalized supported membrane microarrays. This enables characterization of the full-length SOS protein, which has not previously been studied in reconstitution due to difficulties in purification. Our measurements on the full-length protein reveal a distinct role of the C-terminal proline-rich (PR) domain to obstruct the engagement of allosteric Ras independently of the well-known N-terminal domain autoinhibition. This inhibitory role of the PR domain limits Grb2-independent recruitment of SOS to the membrane through binding of Ras·GTP in the SOS allosteric binding site. More generally, this assay strategy enables characterization of the functional behaviour of GEFs with single-molecule precision but without the need for purification.
- Published
- 2017
- Full Text
- View/download PDF
49. Click-Chemistry Based High Throughput Screening Platform for Modulators of Ras Palmitoylation.
- Author
-
Ganesan L, Shieh P, Bertozzi CR, and Levental I
- Subjects
- Click Chemistry, Drug Evaluation, Preclinical, Proto-Oncogene Proteins c-fyn pharmacology, ras Proteins chemistry, ras Proteins pharmacokinetics, High-Throughput Screening Assays methods, Lipoylation, ras Proteins antagonists & inhibitors
- Abstract
Palmitoylation is a widespread, reversible lipid modification that has been implicated in regulating a variety of cellular processes. Approximately one thousand proteins are annotated as being palmitoylated, and for some of these, including several oncogenes of the Ras and Src families, palmitoylation is indispensable for protein function. Despite this wealth of disease-relevant targets, there are currently few effective pharmacological tools to interfere with protein palmitoylation. One reason for this lack of development is the dearth of assays to efficiently screen for small molecular inhibitors of palmitoylation. To address this shortcoming, we have developed a robust, high-throughput compatible, click chemistry-based approach to identify small molecules that interfere with the palmitoylation of Ras, a high value therapeutic target that is mutated in up to a third of human cancers. This assay design shows excellent performance in 384-well format and is sensitive to known, non-specific palmitoylation inhibitors. Further, we demonstrate an ideal counter-screening strategy, which relies on a target peptide from an unrelated protein, the Src-family kinase Fyn. The screening approach described here provides an integrated platform to identify specific modulators of palmitoylated proteins, demonstrated here for Ras and Fyn, but potentially applicable to pharmaceutical targets involved in a variety of human diseases.
- Published
- 2017
- Full Text
- View/download PDF
50. Computational and biochemical characterization of two partially overlapping interfaces and multiple weak-affinity K-Ras dimers.
- Author
-
Prakash P, Sayyed-Ahmad A, Cho KJ, Dolino DM, Chen W, Li H, Grant BJ, Hancock JF, and Gorfe AA
- Subjects
- Computational Biology, Models, Molecular, Protein Binding, Protein Interaction Domains and Motifs, Protein Multimerization, ras Proteins chemistry, ras Proteins genetics
- Abstract
Recent studies found that membrane-bound K-Ras dimers are important for biological function. However, the structure and thermodynamic stability of these complexes remained unknown because they are hard to probe by conventional approaches. Combining data from a wide range of computational and experimental approaches, here we describe the structure, dynamics, energetics and mechanism of assembly of multiple K-Ras dimers. Utilizing a range of techniques for the detection of reactive surfaces, protein-protein docking and molecular simulations, we found that two largely polar and partially overlapping surfaces underlie the formation of multiple K-Ras dimers. For validation we used mutagenesis, electron microscopy and biochemical assays under non-denaturing conditions. We show that partial disruption of a predicted interface through charge reversal mutation of apposed residues reduces oligomerization while introduction of cysteines at these positions enhanced dimerization likely through the formation of an intermolecular disulfide bond. Free energy calculations indicated that K-Ras dimerization involves direct but weak protein-protein interactions in solution, consistent with the notion that dimerization is facilitated by membrane binding. Taken together, our atomically detailed analyses provide unique mechanistic insights into K-Ras dimer formation and membrane organization as well as the conformational fluctuations and equilibrium thermodynamics underlying these processes.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.