Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Biochem Soc Trans ; 52(3): 1061-1069, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38695730

ABSTRACT

The RAF kinases are required for signal transduction through the RAS-RAF-MEK-ERK pathway, and their activity is frequently up-regulated in human cancer and the RASopathy developmental syndromes. Due to their complex activation process, developing drugs that effectively target RAF function has been a challenging endeavor, highlighting the need for a more detailed understanding of RAF regulation. This review will focus on recent structural and biochemical studies that have provided 'snapshots' into the RAF regulatory cycle, revealing structures of the autoinhibited BRAF monomer, active BRAF and CRAF homodimers, as well as HSP90/CDC37 chaperone complexes containing CRAF or BRAFV600E. In addition, we will describe the insights obtained regarding how BRAF transitions between its regulatory states and examine the roles that various BRAF domains and 14-3-3 dimers play in both maintaining BRAF as an autoinhibited monomer and in facilitating its transition to an active dimer. We will also address the function of the HSP90/CDC37 chaperone complex in stabilizing the protein levels of CRAF and certain oncogenic BRAF mutants, and in serving as a platform for RAF dephosphorylation mediated by the PP5 protein phosphatase. Finally, we will discuss the regulatory differences observed between BRAF and CRAF and how these differences impact the function of BRAF and CRAF as drivers of human disease.


Subject(s)
HSP90 Heat-Shock Proteins , Proto-Oncogene Proteins B-raf , Humans , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/chemistry , Protein Multimerization , raf Kinases/metabolism , raf Kinases/chemistry , Animals , Chaperonins/metabolism , Chaperonins/chemistry , Signal Transduction , 14-3-3 Proteins/metabolism , 14-3-3 Proteins/chemistry , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/genetics , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/chemistry , Models, Molecular
2.
STAR Protoc ; 4(3): 102461, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37590148

ABSTRACT

BRAF is frequently activated via mutation in human cancer and the RASopathy syndromes; however, for BRAF activation to occur, autoinhibitory interactions between the regulatory and catalytic domains must be relieved. Here, we present a proximity-based NanoBRET (bioluminescence resonance energy transfer) assay for real-time measurement of BRAF autoinhibition in live cells. We describe steps for seeding, transfecting, and replating cells. We then detail procedures for reading the NanoBRET emissions and confirming protein expression. For complete details on the use and execution of this protocol, please refer to Spencer-Smith et al. (2022).1.


Subject(s)
Biological Assay , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Mutation
3.
Mol Cell ; 82(22): 4262-4276.e5, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36347258

ABSTRACT

BRAF is frequently mutated in human cancer and the RASopathy syndromes, with RASopathy mutations often observed in the cysteine-rich domain (CRD). Although the CRD participates in phosphatidylserine (PS) binding, the RAS-RAF interaction, and RAF autoinhibition, the impact of these activities on RAF function in normal and disease states is not well characterized. Here, we analyze a panel of CRD mutations and show that they increase BRAF activity by relieving autoinhibition and/or enhancing PS binding, with relief of autoinhibition being the major factor determining mutation severity. Further, we show that CRD-mediated autoinhibition prevents the constitutive plasma membrane localization of BRAF that causes increased RAS-dependent and RAS-independent function. Comparison of the BRAF- and CRAF-CRDs also indicates that the BRAF-CRD is a stronger mediator of autoinhibition and PS binding, and given the increased catalytic activity of BRAF, our studies reveal a more critical role for CRD-mediated autoinhibition in BRAF regulation.


Subject(s)
Cysteine , Proto-Oncogene Proteins B-raf , Humans , Cysteine/genetics , Proto-Oncogene Proteins B-raf/genetics , Protein Domains , Mutation , Syndrome
4.
J Nat Prod ; 85(6): 1603-1616, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35696348

ABSTRACT

Seven new peptaibols named tolypocladamides A-G have been isolated from an extract of the fungus Tolypocladium inflatum, which inhibits the interaction between Raf and oncogenic Ras in a cell-based high-throughput screening assay. Each peptaibol contains 11 amino acid residues, an octanoyl or decanoyl fatty acid chain at the N-terminus, and a leucinol moiety at the C-terminus. The peptaibol sequences were elucidated on the basis of 2D NMR and mass spectral fragmentation analyses. Amino acid configurations were determined by advanced Marfey's analyses. Tolypocladamides A-G caused significant inhibition of Ras/Raf interactions with IC50 values ranging from 0.5 to 5.0 µM in a nanobioluminescence resonance energy transfer (NanoBRET) assay; however, no interactions were observed in a surface plasmon resonance assay for binding of the compounds to wild type or G12D mutant Ras constructs or to the Ras binding domain of Raf. NCI 60 cell line testing was also conducted, and little panel selectivity was observed.


Subject(s)
Antineoplastic Agents , Hypocreales , Amino Acids/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Hypocreales/chemistry , Peptaibols/pharmacology
5.
Nat Commun ; 13(1): 3153, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672316

ABSTRACT

A distinct profile of NRAS mutants is observed in each tumor type. It is unclear whether these profiles are determined by mutagenic events or functional differences between NRAS oncoproteins. Here, we establish functional hallmarks of NRAS mutants enriched in human melanoma. We generate eight conditional, knock-in mouse models and show that rare melanoma mutants (NRAS G12D, G13D, G13R, Q61H, and Q61P) are poor drivers of spontaneous melanoma formation, whereas common melanoma mutants (NRAS Q61R, Q61K, or Q61L) induce rapid tumor onset with high penetrance. Molecular dynamics simulations, combined with cell-based protein-protein interaction studies, reveal that melanomagenic NRAS mutants form intramolecular contacts that enhance BRAF binding affinity, BRAF-CRAF heterodimer formation, and MAPK > ERK signaling. Along with the allelic series of conditional mouse models we describe, these results establish a mechanistic basis for the enrichment of specific NRAS mutants in human melanoma.


Subject(s)
Melanoma , Monomeric GTP-Binding Proteins/standards , Skin Neoplasms , Animals , Disease Models, Animal , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Mice , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction/genetics , Skin Neoplasms/genetics
6.
Mol Cell ; 82(5): 950-968.e14, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35202574

ABSTRACT

A unifying feature of the RAS superfamily is a conserved GTPase cycle by which these proteins transition between active and inactive states. We demonstrate that autophosphorylation of some GTPases is an intrinsic regulatory mechanism that reduces nucleotide hydrolysis and enhances nucleotide exchange, altering the on/off switch that forms the basis for their signaling functions. Using X-ray crystallography, nuclear magnetic resonance spectroscopy, binding assays, and molecular dynamics on autophosphorylated mutants of H-RAS and K-RAS, we show that phosphoryl transfer from GTP requires dynamic movement of the switch II region and that autophosphorylation promotes nucleotide exchange by opening the active site and extracting the stabilizing Mg2+. Finally, we demonstrate that autophosphorylated K-RAS exhibits altered effector interactions, including a reduced affinity for RAF proteins in mammalian cells. Thus, autophosphorylation leads to altered active site dynamics and effector interaction properties, creating a pool of GTPases that are functionally distinct from their non-phosphorylated counterparts.


Subject(s)
GTP Phosphohydrolases , Signal Transduction , Animals , Crystallography, X-Ray , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/metabolism , Mammals/metabolism , Nucleotides , Proteins
7.
Cancer Discov ; 12(4): 899-912, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35046094

ABSTRACT

The RAS GTPases are frequently mutated in human cancer, with KRAS being the predominant tumor driver. For many years, it has been known that the structure and function of RAS are integrally linked, as structural changes induced by GTP binding or mutational events determine the ability of RAS to interact with regulators and effectors. Recently, a wealth of information has emerged from structures of specific KRAS mutants and from structures of multiprotein complexes containing RAS and/or RAF, an essential effector of RAS. These structures provide key insights regarding RAS and RAF regulation as well as promising new strategies for therapeutic intervention. SIGNIFICANCE: The RAS GTPases are major drivers of tumorigenesis, and for RAS proteins to exert their full oncogenic potential, they must interact with the RAF kinases to initiate ERK cascade signaling. Although binding to RAS is typically a prerequisite for RAF to become an activated kinase, determining the molecular mechanisms by which this interaction results in RAF activation has been a challenging task. A major advance in understanding this process and RAF regulation has come from recent structural studies of various RAS and RAF multiprotein signaling complexes, revealing new avenues for drug discovery.


Subject(s)
raf Kinases , ras Proteins , Humans , MAP Kinase Signaling System , Oncogenes , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/metabolism
8.
Nat Commun ; 13(1): 486, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35078985

ABSTRACT

RAF kinases are essential effectors of RAS, but how RAS binding initiates the conformational changes needed for autoinhibited RAF monomers to form active dimers has remained unclear. Here, we present cryo-electron microscopy structures of full-length BRAF complexes derived from mammalian cells: autoinhibited, monomeric BRAF:14-3-32:MEK and BRAF:14-3-32 complexes, and an inhibitor-bound, dimeric BRAF2:14-3-32 complex, at 3.7, 4.1, and 3.9 Å resolution, respectively. In both autoinhibited, monomeric structures, the RAS binding domain (RBD) of BRAF is resolved, revealing that the RBD forms an extensive contact interface with the 14-3-3 protomer bound to the BRAF C-terminal site and that key basic residues required for RBD-RAS binding are exposed. Moreover, through structure-guided mutational studies, our findings indicate that RAS-RAF binding is a dynamic process and that RBD residues at the center of the RBD:14-3-3 interface have a dual function, first contributing to RAF autoinhibition and then to the full spectrum of RAS-RBD interactions.


Subject(s)
Cryoelectron Microscopy/methods , Mutation , Neoplasms/pathology , Protein Multimerization , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/metabolism , Animals , Cell Line , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Protein Conformation , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/chemistry
9.
Mol Cancer Ther ; 20(9): 1743-1754, 2021 09.
Article in English | MEDLINE | ID: mdl-34158349

ABSTRACT

Activating mutations in RAS are found in approximately 30% of human cancers, resulting in the delivery of a persistent signal to critical downstream effectors that drive tumorigenesis. RAS-driven malignancies respond poorly to conventional cancer treatments and inhibitors that target RAS directly are limited; therefore, the identification of new strategies and/or drugs to disrupt RAS signaling in tumor cells remains a pressing therapeutic need. Taking advantage of the live-cell bioluminescence resonance energy transfer (BRET) methodology, we describe the development of a NanoBRET screening platform to identify compounds that modulate binding between activated KRAS and the CRAF kinase, an essential effector of RAS that initiates ERK cascade signaling. Using this strategy, libraries containing synthetic compounds, targeted inhibitors, purified natural products, and natural product extracts were evaluated. These efforts resulted in the identification of compounds that inhibit RAS/RAF binding and in turn suppress RAS-driven ERK activation, but also compounds that have the deleterious effect of enhancing the interaction to upregulate pathway signaling. Among the inhibitor hits identified, the majority were compounds derived from natural products, including ones reported to alter KRAS nanoclustering (ophiobolin A), to impact RAF function (HSP90 inhibitors and ROS inducers) as well as some with unknown targets and activities. These findings demonstrate the potential for this screening platform in natural product drug discovery and in the development of new therapeutic agents to target dysregulated RAS signaling in human disease states such as cancer.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Fibroblasts/drug effects , High-Throughput Screening Assays/methods , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , ras Proteins/agonists , ras Proteins/antagonists & inhibitors , Animals , Fibroblasts/metabolism , Humans , Ligands , Nanotechnology/methods , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism
10.
Cancer Discov ; 11(6): 1411-1423, 2021 06.
Article in English | MEDLINE | ID: mdl-33495197

ABSTRACT

Lung squamous cell carcinoma (LSCC) is the second most prevalent type of lung cancer. Despite extensive genomic characterization, no targeted therapies are approved for the treatment of LSCC. Distal amplification of the 3q chromosome is the most frequent genomic alteration in LSCC, and there is an urgent need to identify efficacious druggable targets within this amplicon. We identify the protein kinase TNIK as a therapeutic target in LSCC. TNIK is amplified in approximately 50% of LSCC cases. TNIK genetic depletion or pharmacologic inhibition reduces the growth of LSCC cells in vitro and in vivo. In addition, TNIK inhibition showed antitumor activity and increased apoptosis in established LSCC patient-derived xenografts. Mechanistically, we identified the tumor suppressor Merlin/NF2 as a novel TNIK substrate and showed that TNIK and Merlin are required for the activation of focal adhesion kinase. In conclusion, our data identify targeting TNIK as a potential therapeutic strategy in LSCC. SIGNIFICANCE: Targeted therapies have not yet been approved for the treatment of LSCC, due to lack of identification of actionable cancer drivers. We define TNIK catalytic activity as essential for maintaining LSCC viability and validate the antitumor efficacy of TNIK inhibition in preclinical models of LSCC.This article is highlighted in the In This Issue feature, p. 1307.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor/drug effects , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Mice , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics
11.
Mol Cancer Ther ; 20(2): 307-319, 2021 02.
Article in English | MEDLINE | ID: mdl-33158997

ABSTRACT

Relapsed pediatric rhabdomyosarcomas (RMS) and neuroblastomas (NBs) have a poor prognosis despite multimodality therapy. In addition, the current standard of care for these cancers includes vinca alkaloids that have severe toxicity profiles, further underscoring the need for novel therapies for these malignancies. Here, we show that the small-molecule rigosertib inhibits the growth of RMS and NB cell lines by arresting cells in mitosis, which leads to cell death. Our data indicate that rigosertib, like the vinca alkaloids, exerts its effects mainly by interfering with mitotic spindle assembly. Although rigosertib has the ability to inhibit oncogenic RAS signaling, we provide evidence that rigosertib does not induce cell death through inhibition of the RAS pathway in RAS-mutated RMS and NB cells. However, the combination of rigosertib and the MEK inhibitor trametinib, which has efficacy in RAS-mutated tumors, synergistically inhibits the growth of an RMS cell line, suggesting a new avenue for combination therapy. Importantly, rigosertib treatment delays tumor growth and prolongs survival in a xenograft model of RMS. In conclusion, rigosertib, through its impact on the mitotic spindle, represents a potential therapeutic for RMS.


Subject(s)
Glycine/analogs & derivatives , Neuroblastoma/drug therapy , Rhabdomyosarcoma/drug therapy , Spindle Apparatus/metabolism , Sulfones/therapeutic use , Apoptosis , Glycine/pharmacology , Glycine/therapeutic use , Humans , Sulfones/pharmacology
12.
J Nat Prod ; 83(4): 1288-1294, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32191460

ABSTRACT

Two new cyclic depsipeptides named swinhopeptolides A (1) and B (2) have been isolated from the marine sponge Theonella swinhoei cf. verrucosa, collected from Papua New Guinea. They each contain 11 diverse amino acid residues and 13-carbon polyketide moieties attached at the N-terminus. Compounds 1 and 2 each exist as two conformers in DMSO-d6 due to cis/trans isomerism of the proline residue, and their structures were successfully assigned by extensive NMR analyses complemented by chemical degradation and derivatization studies. Swinhopeptolide B (2) contains a previously undescribed 2,6,8-trimethyldeca-(2E,4E,6E)-trienoic acid moiety N-linked to a terminal serine residue. Swinhopeptolides A (1) and B (2) showed significant inhibition of the Ras/Raf signaling pathway with IC50 values of 5.8 and 8.5 µM, respectively.


Subject(s)
Depsipeptides/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Theonella/chemistry , ras Proteins/antagonists & inhibitors , Amino Acids/chemistry , Animals , Depsipeptides/chemistry , Depsipeptides/isolation & purification , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Papua New Guinea , Porifera/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction/drug effects , ras Proteins/metabolism
13.
Am J Med Genet A ; 182(4): 866-876, 2020 04.
Article in English | MEDLINE | ID: mdl-31913576

ABSTRACT

RASopathies caused by germline pathogenic variants in genes that encode RAS pathway proteins. These disorders include neurofibromatosis type 1 (NF1), Noonan syndrome (NS), cardiofaciocutaneous syndrome (CFC), and Costello syndrome (CS), and others. RASopathies are characterized by heterogenous manifestations, including congenital heart disease, failure to thrive, and increased risk of cancers. Previous work led by the NCI Pediatric Oncology Branch has altered the natural course of one of the key manifestations of the RASopathy NF1. Through the conduct of a longitudinal cohort study and early phase clinical trials, the MEK inhibitor selumetinib was identified as the first active therapy for the NF1-related peripheral nerve sheath tumors called plexiform neurofibromas (PNs). As a result, selumetinib was granted breakthrough therapy designation by the FDA for the treatment of PN. Other RASopathy manifestations may also benefit from RAS targeted therapies. The overall goal of Advancing RAS/RASopathy Therapies (ART), a new NCI initiative, is to develop effective therapies and prevention strategies for the clinical manifestations of the non-NF1 RASopathies and for tumors characterized by somatic RAS mutations. This report reflects discussions from a February 2019 initiation meeting for this project, which had broad international collaboration from basic and clinical researchers and patient advocates.


Subject(s)
Costello Syndrome/therapy , Ectodermal Dysplasia/therapy , Failure to Thrive/therapy , Heart Defects, Congenital/therapy , Molecular Targeted Therapy , Mutation , Neurofibromatosis 1/therapy , Noonan Syndrome/therapy , ras Proteins/antagonists & inhibitors , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Costello Syndrome/genetics , Costello Syndrome/pathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Facies , Failure to Thrive/genetics , Failure to Thrive/pathology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Humans , Intersectoral Collaboration , National Cancer Institute (U.S.) , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Research Report , Signal Transduction , United States , ras Proteins/genetics
14.
Mol Cell ; 76(6): 872-884.e5, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31606273

ABSTRACT

The Ras GTPases are frequently mutated in human cancer, and, although the Raf kinases are essential effectors of Ras signaling, the tumorigenic properties of specific Ras-Raf complexes are not well characterized. Here, we examine the ability of individual Ras and Raf proteins to interact in live cells using bioluminescence resonance energy transfer (BRET) technology. We find that C-Raf binds all mutant Ras proteins with high affinity, whereas B-Raf exhibits a striking preference for mutant K-Ras. This selectivity is mediated by the acidic, N-terminal segment of B-Raf and requires the K-Ras polybasic region for high-affinity binding. In addition, we find that C-Raf is critical for mutant H-Ras-driven signaling and that events stabilizing B-Raf/C-Raf dimerization, such as Raf inhibitor treatment or certain B-Raf mutations, can allow mutant H-Ras to engage B-Raf with increased affinity to promote tumorigenesis, thus revealing a previously unappreciated role for C-Raf in potentiating B-Raf function.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Neoplasms/enzymology , raf Kinases/metabolism , ras Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mutation , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/genetics , Spheroids, Cellular , raf Kinases/genetics , ras Proteins/genetics
15.
Dev Cell ; 50(2): 229-246.e7, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31204173

ABSTRACT

Serum starvation stimulates cilia growth in cultured cells, yet serum factors associated with ciliogenesis are unknown. Previously, we showed that starvation induces rapid Rab11-dependent vesicular trafficking of Rabin8, a Rab8 guanine-nucleotide exchange factor (GEF), to the mother centriole, leading to Rab8 activation and cilium growth. Here, we demonstrate that through the LPA receptor 1 (LPAR1), serum lysophosphatidic acid (LPA) inhibits Rab11a-Rabin8 interaction and ciliogenesis. LPA/LPAR1 regulates ciliogenesis initiation via downstream PI3K/Akt activation, independent of effects on cell cycle. Akt stabilizes Rab11a binding to its effector, WDR44, and a WDR44-pAkt-phosphomimetic mutant blocks ciliogenesis. WDR44 depletion promotes Rabin8 preciliary trafficking and ciliogenesis-initiating events at the mother centriole. Our work suggests disruption of Akt signaling causes a switch from Rab11-WDR44 to the ciliogenic Rab11-FIP3-Rabin8 complex. Finally, we demonstrate that Akt regulates downstream ciliogenesis processes associated with Rab8-dependent cilia growth. Together, this study uncovers a mechanism whereby serum mitogen signaling regulates Rabin8 preciliary trafficking and ciliogenesis initiation.


Subject(s)
Cilia/physiology , Gene Expression Regulation , I-kappa B Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Humans , I-kappa B Kinase/genetics , Phosphatidylinositol 3-Kinases/genetics , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Zebrafish , rab GTP-Binding Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 116(9): 3536-3545, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808747

ABSTRACT

Collective cell migration is required for normal embryonic development and contributes to various biological processes, including wound healing and cancer cell invasion. The M-Ras GTPase and its effector, the Shoc2 scaffold, are proteins mutated in the developmental RASopathy Noonan syndrome, and, here, we report that activated M-Ras recruits Shoc2 to cell surface junctions where M-Ras/Shoc2 signaling contributes to the dynamic regulation of cell-cell junction turnover required for collective cell migration. MCF10A cells expressing the dominant-inhibitory M-RasS27N variant or those lacking Shoc2 exhibited reduced junction turnover and were unable to migrate effectively as a group. Through further depletion/reconstitution studies, we found that M-Ras/Shoc2 signaling contributes to junction turnover by modulating the E-cadherin/p120-catenin interaction and, in turn, the junctional expression of E-cadherin. The regulatory effect of the M-Ras/Shoc2 complex was mediated at least in part through the phosphoregulation of p120-catenin and required downstream ERK cascade activation. Strikingly, cells rescued with the Noonan-associated, myristoylated-Shoc2 mutant (Myr-Shoc2) displayed a gain-of-function (GOF) phenotype, with the cells exhibiting increased junction turnover and reduced E-cadherin/p120-catenin binding and migrating as a faster but less cohesive group. Consistent with these results, Noonan-associated C-Raf mutants that bypass the need for M-Ras/Shoc2 signaling exhibited a similar GOF phenotype when expressed in Shoc2-depleted MCF10A cells. Finally, expression of the Noonan-associated Myr-Shoc2 or C-Raf mutants, but not their WT counterparts, induced gastrulation defects indicative of aberrant cell migration in zebrafish embryos, further demonstrating the function of the M-Ras/Shoc2/ERK cascade signaling axis in the dynamic control of coordinated cell movement.


Subject(s)
Cell Adhesion/genetics , Cell Movement/genetics , Embryonic Development/genetics , Intracellular Signaling Peptides and Proteins/genetics , Monomeric GTP-Binding Proteins/genetics , Animals , Cadherins/genetics , Gain of Function Mutation/genetics , Gastrulation/genetics , Humans , MAP Kinase Signaling System/genetics , Noonan Syndrome/genetics , Noonan Syndrome/physiopathology , Protein Binding , Zebrafish/genetics
17.
Article in English | MEDLINE | ID: mdl-29358316

ABSTRACT

The extracellular signal-regulated kinase (ERK) cascade comprised of the Raf, MEK, and ERK protein kinases constitutes a key effector cascade used by the Ras GTPases to relay signals regulating cell growth, survival, proliferation, and differentiation. Of the ERK cascade components, the regulation of the Raf kinases is by far the most complex, involving changes in subcellular localization, protein and lipid interactions, as well as alterations in the Raf phosphorylation state. The Raf kinases interact directly with active, membrane-localized Ras, and this interaction is often the first step in the Raf activation process, which ultimately results in ERK activation and the downstream phosphorylation of cellular targets that will specify a particular biological response. Here, we will examine our current understanding of how Ras promotes Raf activation, focusing on the molecular mechanisms that contribute to the Raf activation/inactivation cycle.


Subject(s)
MAP Kinase Signaling System , raf Kinases/metabolism , Animals , Cell Proliferation , Dimerization , Germ-Line Mutation , Humans , Signal Transduction
18.
Front Chem ; 6: 434, 2018.
Article in English | MEDLINE | ID: mdl-30324102

ABSTRACT

In the present study, a series of novel madecassic acid derivatives was synthesized and screened against the National Cancer Institute's 60 human cancer cell line panel. Among them, compounds 5, 12, and 17 displayed potent and highly differential antiproliferative activity against 80% of the tumor cells harboring the B-RafV600E mutation within the nanomolar range. Structure-activity analysis revealed that a 5-membered A ring containing an α,ß-unsaturated aldehyde substituted at C-23 with a 2-furoyl group seems to be crucial to produce this particular growth inhibition signature. In silico analysis of the cytotoxicity pattern of these compounds identified two highly correlated clinically approved drugs with known B-RafV600E inhibitory activity. Follow-up analysis revealed inhibition of the ERK signaling pathway through the reduction of cellular Raf protein levels is a key mechanism of action of these compounds. In particular, 17 was the most potent compound in suppressing tumor growth of B-RafV600E-mutant cell lines and displayed the highest reduction of Raf protein levels among the tested compounds. Taken together, this study revealed that modifications of madecassic acid structure can provide molecules with potent anticancer activity against cell lines harboring the clinically relevant B-RafV600E mutation, with compound 17 identified as a promising lead for the development of new anticancer drugs.

19.
Sci Signal ; 11(546)2018 09 04.
Article in English | MEDLINE | ID: mdl-30181243

ABSTRACT

About a third of tumors have activating mutations in HRAS, NRAS, or KRAS, genes encoding guanosine triphosphatases (GTPases) of the RAS family. In these tumors, wild-type RAS cooperates with mutant RAS to promote downstream effector activation and cell proliferation and transformation, suggesting that upstream activators of wild-type RAS are important modulators of mutant RAS-driven oncogenesis. The guanine nucleotide exchange factor (GEF) SOS1 mediates KRAS-driven proliferation, but little is understood about the role of SOS2. We found that RAS family members have a hierarchical requirement for the expression and activity of SOS2 to drive cellular transformation. In mouse embryonic fibroblasts (MEFs), SOS2 critically mediated mutant KRAS-driven, but not HRAS-driven, transformation. Sos2 deletion reduced epidermal growth factor (EGF)-dependent activation of wild-type HRAS and phosphorylation of the kinase AKT in cells expressing mutant RAS isoforms. Assays using pharmacological inhibitors revealed a hierarchical requirement for signaling by phosphoinositide 3-kinase (PI3K) in promoting RAS-driven cellular transformation that mirrored the requirement for SOS2. KRAS-driven transformation required the GEF activity of SOS2 and was restored in Sos2-/- MEFs by expression of constitutively activated PI3K. Finally, CRISPR/Cas9-mediated deletion of SOS2 reduced EGF-stimulated AKT phosphorylation and synergized with MEK inhibition to revert the transformed phenotype of human KRAS mutant pancreatic and lung tumor cells. These results indicate that SOS2-dependent PI3K signaling mediates mutant KRAS-driven transformation, revealing therapeutic targets in KRAS-driven cancers. Our data also reveal the importance of three-dimensional culture systems in investigating the mediators of mutant KRAS.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblasts/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Son of Sevenless Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , Humans , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/genetics , Son of Sevenless Proteins/genetics
20.
J Nat Prod ; 81(7): 1666-1672, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29979591

ABSTRACT

Six new macrophilone-type pyrroloiminoquines were isolated and identified from an extract of the marine hydroid Macrorhynchia philippina. The proton-deficient and heteroatom-rich structures of macrophilones B-G (2-7) were elucidated by spectroscopic analysis and comparison of their data with those of the previously reported metabolite macrophilone A (1). Compounds 1-7 are the first pyrroloiminoquines to be reported from a hydroid. The macrophilones were shown to inhibit the enzymatic conjugation of SUMO to peptide substrates, and macrophilones A (1) and C (3) exhibit potent and selective cytotoxic properties in the NCI-60 anticancer screen. Bioinformatic analysis revealed a close association of the cytotoxicity profiles of 1 and 3 with two known B-Raf kinase inhibitory drugs. While compounds 1 and 3 showed no kinase inhibitory activity, they resulted in a dramatic decrease in cellular protein levels of selected components of the ERK signal cascade. As such, the chemical scaffold of the macrophilones could provide small-molecule therapeutic leads that target the ERK signal transduction pathway.


Subject(s)
Hydrozoa/chemistry , MAP Kinase Signaling System/drug effects , Pyrroloiminoquinones/isolation & purification , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Pyrroloiminoquinones/pharmacology , Sumoylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...