Recent Insights into the Regulation and Structure of RAS Membrane Signaling Complexes—Emerging Drug Targeting Opportunities

A special issue of Biomolecules (ISSN 2218-273X). This special issue belongs to the section "Molecular Biology".

Deadline for manuscript submissions: closed (28 February 2022) | Viewed by 33209

Special Issue Editor


E-Mail Website
Guest Editor
University of Luxembourg
Interests: Ras; nanocluster; drug development; FRET; Raf; biosensors; RASopathy; cancer stem cells; galectin; PDEdelta

Special Issue Information

Dear Colleagues,

The basic architecture of the Ras signaling network has been known for many years. However, given the difficulties in resolving structures in the membrane context, we still have significant knowledge gaps in that area of Ras biology. For instance, we actually do not know the structural details of how Ras activates Raf exactly. Such knowledge is very important, as it may have predicted the clinically relevant paradoxical activation of MAPK-signaling with Raf-dimer stabilizing inhibitors, such as vemurafenib.

Currently, the first direct Ras inhibitors are being tested in the clinic and will help us to understand the therapeutic potential of inhibiting one of the first and primary cancer drug targets. In the wake of these drug development efforts, a significant amount of new Ras research is providing us with unprecedented details of how the molecular machinery of Ras signaling is functioning. We now know that Ras is organized in di/oligomeric nanoclusters of the membrane that are necessary for its signal transmission. Conformational reorientation of Ras in the membrane seems to guide effector interactions, and allosteric communication routes between effectors and Ras are emerging. Moreover, we know that trafficking chaperones aid Ras in dynamically populating subcellular membrane compartments, where Ras activity is little investigated.

This refined understanding of Ras biology was enabled in particular by improved computational, biophysical, and microscopic tools that allowed us to investigate Ras signaling complexes in their native membrane environment. Importantly, new molecular mechanistic insight can suggest novel entry points for future drug targeting. In addition, they help us to understand the mechanistic background of disease-associated mutations within the Ras/MAPK pathway.

Prof. Dr. Daniel Abankwa
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomolecules is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Ras nanocluster
  • Ras dimer
  • structure
  • simulations
  • Ras membrane conformation
  • Ras membrane trafficking
  • Raf
  • Mek
  • Sos
  • inhibitor
  • drug targeting
  • cancer
  • RASopathy

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

19 pages, 5659 KiB  
Article
Nanoscopic Spatial Association between Ras and Phosphatidylserine on the Cell Membrane Studied with Multicolor Super Resolution Microscopy
by Anna M. Koester, Kai Tao, Malwina Szczepaniak, Matthew J. Rames and Xiaolin Nan
Biomolecules 2022, 12(8), 1033; https://doi.org/10.3390/biom12081033 - 26 Jul 2022
Cited by 5 | Viewed by 2642
Abstract
Recent work suggests that Ras small GTPases interact with the anionic lipid phosphatidylserine (PS) in an isoform-specific manner, with direct implications for their biological functions. Studies on PS-Ras associations in cells, however, have relied on immuno-EM imaging of membrane sheets. To study their [...] Read more.
Recent work suggests that Ras small GTPases interact with the anionic lipid phosphatidylserine (PS) in an isoform-specific manner, with direct implications for their biological functions. Studies on PS-Ras associations in cells, however, have relied on immuno-EM imaging of membrane sheets. To study their spatial relationships in intact cells, we have combined the use of Lact-C2-GFP, a biosensor for PS, with multicolor super resolution imaging based on DNA-PAINT. At ~20 nm spatial resolution, the resulting super resolution images clearly show the nonuniform molecular distribution of PS on the cell membrane and its co-enrichment with caveolae, as well as with unidentified membrane structures. Two-color imaging followed by spatial analysis shows that KRas-G12D and HRas-G12V both co-enrich with PS in model U2OS cells, confirming previous observations, yet exhibit clear differences in their association patterns. Whereas HRas-G12V is almost always co-enriched with PS, KRas-G12D is strongly co-enriched with PS in about half of the cells, with the other half exhibiting a more moderate association. In addition, perturbations to the actin cytoskeleton differentially impact PS association with the two Ras isoforms. These results suggest that PS-Ras association is context-dependent and demonstrate the utility of multiplexed super resolution imaging in defining the complex interplay between Ras and the membrane. Full article
Show Figures

Figure 1

20 pages, 5740 KiB  
Article
Crystal Structure Reveals the Full Ras–Raf Interface and Advances Mechanistic Understanding of Raf Activation
by Trinity Cookis and Carla Mattos
Biomolecules 2021, 11(7), 996; https://doi.org/10.3390/biom11070996 - 07 Jul 2021
Cited by 28 | Viewed by 4491
Abstract
Ras and Raf-kinase interact through the Ras-binding (RBD) and cysteine-rich domains (CRD) of Raf to signal through the mitogen-activated protein kinase pathway, yet the molecular mechanism leading to Raf activation has remained elusive. We present the 2.8 Å crystal structure of the HRas–CRaf-RBD_CRD [...] Read more.
Ras and Raf-kinase interact through the Ras-binding (RBD) and cysteine-rich domains (CRD) of Raf to signal through the mitogen-activated protein kinase pathway, yet the molecular mechanism leading to Raf activation has remained elusive. We present the 2.8 Å crystal structure of the HRas–CRaf-RBD_CRD complex showing the Ras–Raf interface as a continuous surface on Ras, as seen in the KRas–CRaf-RBD_CRD structure. In molecular dynamics simulations of a Ras dimer model formed through the α4–α5 interface, the CRD is dynamic and located between the two Ras protomers, poised for direct or allosteric modulation of functionally relevant regions of Ras and Raf. We propose a molecular model in which Ras binding is involved in the release of Raf autoinhibition while the Ras–Raf complex dimerizes to promote a platform for signal amplification, with Raf-CRD centrally located to impact regulation and function. Full article
Show Figures

Graphical abstract

12 pages, 1702 KiB  
Article
Elaiophylin Is a Potent Hsp90/ Cdc37 Protein Interface Inhibitor with K-Ras Nanocluster Selectivity
by Farid A. Siddiqui, Vladimir Vukic, Tiina A. Salminen and Daniel Abankwa
Biomolecules 2021, 11(6), 836; https://doi.org/10.3390/biom11060836 - 04 Jun 2021
Cited by 3 | Viewed by 2759
Abstract
The natural product elaiophylin is a macrodiolide with a broad range of biological activities. However, no direct target of elaiophylin in eukaryotes has been described so far, which hinders a systematic explanation of its astonishing activity range. We recently showed that the related [...] Read more.
The natural product elaiophylin is a macrodiolide with a broad range of biological activities. However, no direct target of elaiophylin in eukaryotes has been described so far, which hinders a systematic explanation of its astonishing activity range. We recently showed that the related conglobatin A, a protein–protein interface inhibitor of the interaction between the N-terminus of Hsp90 and its cochaperone Cdc37, blocks cancer stem cell properties by selectively inhibiting K-Ras4B but not H-Ras. Here, we elaborated that elaiophylin likewise disrupts the Hsp90/ Cdc37 interaction, without affecting the ATP-pocket of Hsp90. Similarly to conglobatin A, elaiophylin decreased expression levels of the Hsp90 client HIF1α, a transcription factor with various downstream targets, including galectin-3. Galectin-3 is a nanocluster scaffold of K-Ras, which explains the K-Ras selectivity of Hsp90 inhibitors. In agreement with this K-Ras targeting and the potent effect on other Hsp90 clients, we observed with elaiophylin treatment a submicromolar IC50 for MDA-MB-231 and MIA-PaCa-2 3D spheroid formation. Finally, a strong inhibition of MDA-MB-231 cells grown in the chorioallantoic membrane (CAM) microtumor model was determined. These results suggest that several other macrodiolides may have the Hsp90/ Cdc37 interface as a target site. Full article
Show Figures

Graphical abstract

Review

Jump to: Research, Other

17 pages, 1771 KiB  
Review
Lipid Profiles of RAS Nanoclusters Regulate RAS Function
by Yong Zhou and John F. Hancock
Biomolecules 2021, 11(10), 1439; https://doi.org/10.3390/biom11101439 - 30 Sep 2021
Cited by 13 | Viewed by 2295
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 [...] Read more.
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. Full article
Show Figures

Figure 1

20 pages, 3844 KiB  
Review
RAS Nanoclusters: Dynamic Signaling Platforms Amenable to Therapeutic Intervention
by Que N. Van, Priyanka Prakash, Rebika Shrestha, Trent E. Balius, Thomas J. Turbyville and Andrew G. Stephen
Biomolecules 2021, 11(3), 377; https://doi.org/10.3390/biom11030377 - 03 Mar 2021
Cited by 17 | Viewed by 3841
Abstract
RAS proteins are mutated in approximately 20% of all cancers and are generally associated with poor clinical outcomes. RAS proteins are localized to the plasma membrane and function as molecular switches, turned on by partners that receive extracellular mitogenic signals. In the on-state, [...] Read more.
RAS proteins are mutated in approximately 20% of all cancers and are generally associated with poor clinical outcomes. RAS proteins are localized to the plasma membrane and function as molecular switches, turned on by partners that receive extracellular mitogenic signals. In the on-state, they activate intracellular signal transduction cascades. Membrane-bound RAS molecules segregate into multimers, known as nanoclusters. These nanoclusters, held together through weak protein–protein and protein–lipid associations, are highly dynamic and respond to cellular input signals and fluctuations in the local lipid environment. Disruption of RAS nanoclusters results in downregulation of RAS-mediated mitogenic signaling. In this review, we discuss the propensity of RAS proteins to display clustering behavior and the interfaces that are associated with these assemblies. Strategies to therapeutically disrupt nanocluster formation or the stabilization of signaling incompetent RAS complexes are discussed. Full article
Show Figures

Figure 1

28 pages, 10432 KiB  
Review
The Ins and Outs of RAS Effector Complexes
by Christina Kiel, David Matallanas and Walter Kolch
Biomolecules 2021, 11(2), 236; https://doi.org/10.3390/biom11020236 - 07 Feb 2021
Cited by 27 | Viewed by 5977
Abstract
RAS oncogenes are among the most commonly mutated proteins in human cancers. They regulate a wide range of effector pathways that control cell proliferation, survival, differentiation, migration and metabolic status. Including aberrations in these pathways, RAS-dependent signaling is altered in more than half [...] Read more.
RAS oncogenes are among the most commonly mutated proteins in human cancers. They regulate a wide range of effector pathways that control cell proliferation, survival, differentiation, migration and metabolic status. Including aberrations in these pathways, RAS-dependent signaling is altered in more than half of human cancers. Targeting mutant RAS proteins and their downstream oncogenic signaling pathways has been elusive. However, recent results comprising detailed molecular studies, large scale omics studies and computational modeling have painted a new and more comprehensive portrait of RAS signaling that helps us to understand the intricacies of RAS, how its physiological and pathophysiological functions are regulated, and how we can target them. Here, we review these efforts particularly trying to relate the detailed mechanistic studies with global functional studies. We highlight the importance of computational modeling and data integration to derive an actionable understanding of RAS signaling that will allow us to design new mechanism-based therapies for RAS mutated cancers. Full article
Show Figures

Figure 1

18 pages, 1786 KiB  
Review
Mechanisms of Ras Membrane Organization and Signaling: Ras Rocks Again
by Daniel Abankwa and Alemayehu A. Gorfe
Biomolecules 2020, 10(11), 1522; https://doi.org/10.3390/biom10111522 - 06 Nov 2020
Cited by 29 | Viewed by 4105
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 [...] Read more.
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. Full article
Show Figures

Graphical abstract

Other

Jump to: Research, Review

4 pages, 705 KiB  
Commentary
Spotlight on Accessory Proteins: RTK-RAS-MAPK Modulators as New Therapeutic Targets
by Silke Pudewell and Mohammad Reza Ahmadian
Biomolecules 2021, 11(6), 895; https://doi.org/10.3390/biom11060895 - 16 Jun 2021
Cited by 4 | Viewed by 3181
Abstract
The RTK-RAS-MAPK axis is one of the most extensively studied signaling cascades and is related to the development of both cancers and RASopathies. In the last 30 years, many ideas and approaches have emerged for directly targeting constituent members of this cascade, predominantly [...] Read more.
The RTK-RAS-MAPK axis is one of the most extensively studied signaling cascades and is related to the development of both cancers and RASopathies. In the last 30 years, many ideas and approaches have emerged for directly targeting constituent members of this cascade, predominantly in the context of cancer treatment. These approaches are still insufficient due to undesirable drug toxicity, resistance, and low efficacy. Significant advances have been made in understanding the spatiotemporal features of the constituent members of the RTK-RAS-MAPK axis, which are linked and modulated by many accessory proteins. Given that the majority of such modulators are now emerging as attractive therapeutic targets, a very small number of accessory inhibitors have yet to be discovered. Full article
Show Figures

Figure 1

13 pages, 2502 KiB  
Brief Report
Allosteric Kinase Inhibitors Reshape MEK1 Kinase Activity Conformations in Cells and In Silico
by Jakob Fleischmann, Andreas Feichtner, Louis DeFalco, Valentina Kugler, Selina Schwaighofer, Roland G Huber and Eduard Stefan
Biomolecules 2021, 11(4), 518; https://doi.org/10.3390/biom11040518 - 30 Mar 2021
Cited by 4 | Viewed by 2988
Abstract
Mutations at different stages of the mitogen-activated protein kinase (MAPK) signaling pathway lead to aberrant activation of the involved protein kinase entities. These oncogenic modifications alter signal propagation which converge on the gatekeeper kinases MEK1/2, transmitting the input signal to ERK1/2. Thus, targeted [...] Read more.
Mutations at different stages of the mitogen-activated protein kinase (MAPK) signaling pathway lead to aberrant activation of the involved protein kinase entities. These oncogenic modifications alter signal propagation which converge on the gatekeeper kinases MEK1/2, transmitting the input signal to ERK1/2. Thus, targeted MEK inhibition causes qualitative alterations of carcinogenic MAPK signals. Phosphorylation of the MEK1 activation loop at the positions S218 and S222 by RAF kinases triggers the conformational alignment of MEK’s catalytic pocket to enable ATP-binding and substrate phosphorylation. We have extended a kinase conformation (KinCon) biosensor platform to record MEK1 activity dynamics. In addition to MEK phosphorylation by BRAF, the integration of the phosphorylation-mimetic mutations S218D/S222D triggered opening of the kinase. Structural rearrangement may involve the flexibility of the N terminal MEK1 A-helix. Application of the allosterically acting MEK inhibitors (MEKi) trametinib, cobimentinib, refametinib, and selumetinib converted activated MEK1 KinCon reporters back into a more closed inactive conformation. We confirmed MEK1 KinCon activity dynamics upon drug engagement using the patient-derived melanoma cell line A2058, which harbors the V600E hotspot BRAF mutation. In order to confirm biosensor dynamics, we simulated structure dynamics of MEK1 kinase in the presence and absence of mutations and/or MEKi binding. We observed increased dynamics for the S218D/S222D double mutant particularly in the region of the distal A-helix and alpha-C helix. These data underline that MEK1 KinCon biosensors have the potential to be subjected to MEKi efficacy validations in an intact cell setting. Full article
Show Figures

Figure 1

Back to TopTop