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










Publication year range
1.
Nat Struct Mol Biol ; 29(10): 966-977, 2022 10.
Article in English | MEDLINE | ID: mdl-36175670

ABSTRACT

SHOC2 acts as a strong synthetic lethal interactor with MEK inhibitors in multiple KRAS cancer cell lines. SHOC2 forms a heterotrimeric complex with MRAS and PP1C that is essential for regulating RAF and MAPK-pathway activation by dephosphorylating a specific phosphoserine on RAF kinases. Here we present the high-resolution crystal structure of the SHOC2-MRAS-PP1C (SMP) complex and apo-SHOC2. Our structures reveal that SHOC2, MRAS, and PP1C form a stable ternary complex in which all three proteins synergistically interact with each other. Our results show that dephosphorylation of RAF substrates by PP1C is enhanced upon interacting with SHOC2 and MRAS. The SMP complex forms only when MRAS is in an active state and is dependent on SHOC2 functioning as a scaffolding protein in the complex by bringing PP1C and MRAS together. Our results provide structural insights into the role of the SMP complex in RAF activation and how mutations found in Noonan syndrome enhance complex formation, and reveal new avenues for therapeutic interventions.


Subject(s)
Noonan Syndrome , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Phosphoserine/metabolism , Protein Phosphatase 1 , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/metabolism
2.
PLoS One ; 16(8): e0254697, 2021.
Article in English | MEDLINE | ID: mdl-34424918

ABSTRACT

The PAF complex (PAFC) coordinates transcription elongation and mRNA processing and its CDC73/parafibromin subunit functions as a tumour suppressor. The NF2/Merlin tumour suppressor functions both at the cell cortex and nucleus and is a key mediator of contact inhibition but the molecular mechanisms remain unclear. In this study we have used affinity proteomics to identify novel Merlin interacting proteins and show that Merlin forms a complex with multiple proteins involved in RNA processing including the PAFC and the CHD1 chromatin remodeller. Tumour-derived inactivating mutations in both Merlin and the CDC73 PAFC subunit mutually disrupt their interaction and growth suppression by Merlin requires CDC73. Merlin interacts with the PAFC in a cell density-dependent manner and we identify a role for FAT cadherins in regulating the Merlin-PAFC interaction. Our results suggest that in addition to its function within the Hippo pathway, Merlin is part of a tumour suppressor network regulated by cell-cell adhesion which coordinates post-initiation steps of the transcription cycle of genes mediating contact inhibition.


Subject(s)
Cell Adhesion/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Neurofibromin 2/genetics , Tumor Suppressor Proteins/genetics , Cell Proliferation/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Contact Inhibition/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Neoplasms/pathology , Protein Binding/genetics , Protein Interaction Maps/genetics , Signal Transduction/genetics
3.
Am J Med Genet A ; 182(3): 597-606, 2020 03.
Article in English | MEDLINE | ID: mdl-31825160

ABSTRACT

The RASopathies are a group of genetic disorders that result from germline pathogenic variants affecting RAS-mitogen activated protein kinase (MAPK) pathway genes. RASopathies share RAS/MAPK pathway dysregulation and share phenotypic manifestations affecting numerous organ systems, causing lifelong and at times life-limiting medical complications. RASopathies may benefit from precision medicine approaches. For this reason, the Sixth International RASopathies Symposium focused on exploring precision medicine. This meeting brought together basic science researchers, clinicians, clinician scientists, patient advocates, and representatives from pharmaceutical companies and the National Institutes of Health. Novel RASopathy genes, variants, and animal models were discussed in the context of medication trials and drug development. Attempts to define and measure meaningful endpoints for treatment trials were discussed, as was drug availability to patients after trial completion.


Subject(s)
Genetic Diseases, Inborn/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , ras Proteins/genetics , Genetic Diseases, Inborn/pathology , Germ-Line Mutation/genetics , Humans , Signal Transduction/genetics
4.
Proc Natl Acad Sci U S A ; 116(27): 13330-13339, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31213532

ABSTRACT

Despite the crucial role of RAF kinases in cell signaling and disease, we still lack a complete understanding of their regulation. Heterodimerization of RAF kinases as well as dephosphorylation of a conserved "S259" inhibitory site are important steps for RAF activation but the precise mechanisms and dynamics remain unclear. A ternary complex comprised of SHOC2, MRAS, and PP1 (SHOC2 complex) functions as a RAF S259 holophosphatase and gain-of-function mutations in SHOC2, MRAS, and PP1 that promote complex formation are found in Noonan syndrome. Here we show that SHOC2 complex-mediated S259 RAF dephosphorylation is critically required for growth factor-induced RAF heterodimerization as well as for MEK dissociation from BRAF. We also uncover SHOC2-independent mechanisms of RAF and ERK pathway activation that rely on N-region phosphorylation of CRAF. In DLD-1 cells stimulated with EGF, SHOC2 function is essential for a rapid transient phase of ERK activation, but is not required for a slow, sustained phase that is instead driven by palmitoylated H/N-RAS proteins and CRAF. Whereas redundant SHOC2-dependent and -independent mechanisms of RAF and ERK activation make SHOC2 dispensable for proliferation in 2D, KRAS mutant cells preferentially rely on SHOC2 for ERK signaling under anchorage-independent conditions. Our study highlights a context-dependent contribution of SHOC2 to ERK pathway dynamics that is preferentially engaged by KRAS oncogenic signaling and provides a biochemical framework for selective ERK pathway inhibition by targeting the SHOC2 holophosphatase.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , raf Kinases/chemistry , raf Kinases/metabolism , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Cell Line, Tumor , Gene Editing , Gene Knockout Techniques , Humans , Phosphorylation , Protein Multimerization , ras Proteins/metabolism
5.
Nat Commun ; 10(1): 2532, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182717

ABSTRACT

Targeted inhibition of the ERK-MAPK pathway, upregulated in a majority of human cancers, has been hindered in the clinic by drug resistance and toxicity. The MRAS-SHOC2-PP1 (SHOC2 phosphatase) complex plays a key role in RAF-ERK pathway activation by dephosphorylating a critical inhibitory site on RAF kinases. Here we show that genetic inhibition of SHOC2 suppresses tumorigenic growth in a subset of KRAS-mutant NSCLC cell lines and prominently inhibits tumour development in autochthonous murine KRAS-driven lung cancer models. On the other hand, systemic SHOC2 ablation in adult mice is relatively well tolerated. Furthermore, we show that SHOC2 deletion selectively sensitizes KRAS- and EGFR-mutant NSCLC cells to MEK inhibitors. Mechanistically, SHOC2 deletion prevents MEKi-induced RAF dimerization, leading to more potent and durable ERK pathway suppression that promotes BIM-dependent apoptosis. These results present a rationale for the generation of SHOC2 phosphatase targeted therapies, both as a monotherapy and to widen the therapeutic index of MEK inhibitors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , raf Kinases/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System , Male , Mice, Knockout , Mice, Nude , Mutation , Neoplasm Transplantation , Protein Multimerization , raf Kinases/antagonists & inhibitors , raf Kinases/genetics , ras Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 115(45): E10576-E10585, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30348783

ABSTRACT

Dephosphorylation of the inhibitory "S259" site on RAF kinases (S259 on CRAF, S365 on BRAF) plays a key role in RAF activation. The MRAS GTPase, a close relative of RAS oncoproteins, interacts with SHOC2 and protein phosphatase 1 (PP1) to form a heterotrimeric holoenzyme that dephosphorylates this S259 RAF site. MRAS and SHOC2 function as PP1 regulatory subunits providing the complex with striking specificity against RAF. MRAS also functions as a targeting subunit as membrane localization is required for efficient RAF dephosphorylation and ERK pathway regulation in cells. SHOC2's predicted structure shows remarkable similarities to the A subunit of PP2A, suggesting a case of convergent structural evolution with the PP2A heterotrimer. We have identified multiple regions in SHOC2 involved in complex formation as well as residues in MRAS switch I and the interswitch region that help account for MRAS's unique effector specificity for SHOC2-PP1. MRAS, SHOC2, and PPP1CB are mutated in Noonan syndrome, and we show that syndromic mutations invariably promote complex formation with each other, but not necessarily with other interactors. Thus, Noonan syndrome in individuals with SHOC2, MRAS, or PPPC1B mutations is likely driven at the biochemical level by enhanced ternary complex formation and highlights the crucial role of this phosphatase holoenzyme in RAF S259 dephosphorylation, ERK pathway dynamics, and normal human development.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Noonan Syndrome/metabolism , Protein Phosphatase 1/metabolism , raf Kinases/metabolism , ras Proteins/metabolism , Carrier Proteins , Cell Line , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Models, Molecular , Mutation , Noonan Syndrome/genetics , Phosphorylation , Protein Phosphatase 1/genetics , Sequence Alignment , ras Proteins/genetics
7.
Article in English | MEDLINE | ID: mdl-29311130

ABSTRACT

MRAS is the closest relative to the classical RAS oncoproteins and shares most regulatory and effector interactions. However, it also has unique functions, including its ability to function as a phosphatase regulatory subunit when in complex with SHOC2 and protein phosphatase 1 (PP1). This phosphatase complex regulates a crucial step in the activation cycle of RAF kinases and provides a key coordinate input required for efficient ERK pathway activation and transformation by RAS. MRAS mutations rarely occur in cancer but deregulated expression may play a role in tumorigenesis in some settings. Activating mutations in MRAS (as well as SHOC2 and PP1) do occur in the RASopathy Noonan syndrome, underscoring a key role for MRAS within the RAS-ERK pathway. MRAS also has unique roles in cell migration and differentiation and has properties consistent with a key role in the regulation of cell polarity. Further investigations should shed light on what remains a relatively understudied RAS family member.


Subject(s)
Cell Differentiation/genetics , Cell Movement/genetics , Cell Polarity/genetics , ras Proteins/genetics , Carcinogenesis/genetics , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/genetics , Noonan Syndrome/genetics , Protein Phosphatase 1/metabolism , Signal Transduction , ras Proteins/metabolism
8.
Mol Cell ; 52(5): 679-92, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24211266

ABSTRACT

SHOC2 is mutated in Noonan syndrome and plays a key role in the activation of the ERK-MAPK pathway, which is upregulated in the majority of human cancers. SHOC2 functions as a PP1-regulatory protein and as an effector of MRAS. Here we show that SHOC2 and MRAS form a complex with SCRIB, a polarity protein with tumor suppressor properties. SCRIB functions as a PP1-regulatory protein and antagonizes SHOC2-mediated RAF dephosphorylation through a mechanism involving competition for PP1 molecules within the same macromolecular complex. SHOC2 function is selectively required for the malignant properties of tumor cells with mutant RAS, and both MRAS and SHOC2 play a key role in polarized migration. We propose that MRAS, through its ability to recruit a complex with paradoxical components, coordinates ERK pathway spatiotemporal dynamics with polarity and that this complex plays a key role during tumorigenic growth.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/genetics , Membrane Proteins/genetics , Tumor Suppressor Proteins/genetics , ras Proteins/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line , Cell Movement/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Macromolecular Substances/metabolism , Membrane Proteins/metabolism , Phosphorylation , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Tumor Suppressor Proteins/metabolism , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/metabolism
9.
EMBO Mol Med ; 5(7): 1087-102, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23681745

ABSTRACT

Metastatic spread is the single-most powerful predictor of poor outcome in Ewing sarcoma (ES). Therefore targeting pathways that drive metastasis has tremendous potential to reduce the burden of disease in ES. We previously showed that activation of the ERBB4 tyrosine kinase suppresses anoikis, or detachment-induced cell death, and induces chemoresistance in ES cell lines in vitro. We now show that ERBB4 is transcriptionally overexpressed in ES cell lines derived from chemoresistant or metastatic ES tumours. ERBB4 activates the PI3K-Akt cascade and focal adhesion kinase (FAK), and both pathways contribute to ERBB4-mediated activation of the Rac1 GTPase in vitro and in vivo. ERBB4 augments tumour invasion and metastasis in vivo, and these effects are blocked by ERBB4 knockdown. ERBB4 expression correlates significantly with reduced disease-free survival, and increased expression is observed in metastatic compared to primary patient-matched ES biopsies. Our findings identify a novel ERBB4-PI3K-Akt-FAK-Rac1 pathway associated with aggressive disease in ES. These results predict that therapeutic targeting of ERBB4, alone or in combination with cytotoxic agents, may suppress the metastatic phenotype in ES.


Subject(s)
Bone Neoplasms/pathology , Bone and Bones/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Sarcoma, Ewing/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone and Bones/metabolism , Cell Line, Tumor , Cell Movement , Enzyme Activation , Humans , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4 , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Signal Transduction , Up-Regulation , rac1 GTP-Binding Protein/metabolism
10.
Cancer Res ; 68(19): 8127-36, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829572

ABSTRACT

Mutations in genes functioning in different pathways frequently occur together in the same cancer, whereas mutations in the same pathway tend to be mutually exclusive. However, the majority of colon, breast, and endometrial cancers that possess mutations in PIK3CA, the catalytic subunit p110alpha of phosphatidylinositol 3'-kinase (PI3K), also possess mutations or alterations in genes upstream of PI3K such as Ras, ERBB2/ERBB3, or PTEN. PIK3CA mutations occur almost exclusively in invasive tumors, whereas upstream mutations occur as frequently in early-stage and late-stage tumors, suggesting that PIK3CA mutation is a late-stage event that may augment earlier activation of the PI3K pathway. Consistent with this, we find that levels of p-AKT (Ser(473)) induced by mutant Ras or knockdown of PTEN were dramatically increased by addition of mutant PIK3CA. Soft agar assays revealed that anchorage-independent growth induced by mutant Ras was greatly increased in the presence of mutant PIK3CA. In breast, colon, and endometrial cancers in which the PI3K pathway is activated by a combination of mutant PIK3CA and alterations in Ras, ERBB2/3, or PTEN, signaling to downstream elements such as Akt was mediated exclusively by the p110alpha isoform, rather than a combination of different PI3K isoforms. Our data therefore suggest that in tumors with co-occurring mutations in multiple components of the PI3K pathway, selective inhibition of the alpha isoform of p110 is an attractive therapeutic strategy, especially for late-stage tumors.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Cluster Analysis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, ras/physiology , Humans , Mutation/physiology , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics
11.
Methods Enzymol ; 438: 277-89, 2008.
Article in English | MEDLINE | ID: mdl-18413255

ABSTRACT

Cardio-facio-cutaneous syndrome (CFC) is a sporadic, complex developmental disorder involving characteristic craniofacial features, cardiac defects, ectodermal abnormalities, growth deficiency, hypotonia, and developmental delay. CFC is caused by alteration of activity through the mitogen-activated protein kinase (MAPK) pathway due to heterogeneous de novo germline mutations in B-Raf mutant proteins, MEK1 and MEK2. Approximately 75% of individuals with CFC have mutations in BRAF. In vitro functional studies demonstrate that many of these mutations confer increase activity upon the mutant protein as compared to the wildtype protein. However, as is seen cancer, some of the B-Raf mutant proteins are kinase impaired. Western blot analyses corroborate kinase assays as determined by mutant proteins phosphorylating downstream effectors MEK and ERK. Approximately 25% of individuals with CFC have mutations in either MEK1 or MEK2 that lead to increased MEK kinase activity as judged by increased phosphorylation of its downstream effector ERK. Unlike BRAF, no somatic mutations have ever been identified in MEK genes. The identification of novel germline BRAF and MEK mutations in CFC will help understand the pathophysiology of this syndrome. Furthermore, it will also provide insight to the normal function of B-Raf and MEK, and contribute to the knowledge of the role of the MAPK pathway in cancer. Since the MAPK pathway has been studied intensively in the context of cancer, numerous therapeutics that specifically target this pathway may merit investigation in this population of patients.


Subject(s)
Craniofacial Abnormalities/genetics , Heart Defects, Congenital/genetics , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Proto-Oncogene Proteins B-raf/genetics , Skin Abnormalities/genetics , Amino Acid Substitution , Blotting, Western , Humans , Syndrome , raf Kinases/analysis
12.
Proc Natl Acad Sci U S A ; 103(51): 19290-5, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17158797

ABSTRACT

Binding of Src family kinases to membrane-associated polyoma virus middle T-antigen (PyMT) can result in the phosphorylation of PyMT tyrosine 250, which serves as a docking site for the binding of Shc and subsequent activation of the Raf-MEK-ERK (MAP) kinase cascade. In a screen for PyMT variants that could not activate the ARF tumor suppressor, we isolated a cytoplasmic nontransforming mutant (MTA) that encoded a C-terminal truncated form of the PyMT protein. Surprisingly, MTA was able to strongly activate the MAP kinase pathway in the absence of Src family kinase and Shc binding. Interestingly, the polyoma small T-antigen (PyST), which shares with MTA both partial amino acid sequence homology and cellular location, also activates the MAP kinase cascade. Activation of the MAP kinase cascade by both MTA and PyST has been demonstrated to be PP2A-dependent. Neither MTA nor PyST activate the phosphorylation of AKT. The SV40 small T-antigen, which is similar to PyST in containing a J domain and in binding to the PP2A AC dimer, does not activate the MAP kinase cascade, but does stimulate phosphorylation of AKT in a PP2A-dependent manner. These findings highlight a novel role of PP2A in stimulating the MAP kinase cascade and indicate that the similar polyoma and SV40 small T-antigens influence PP2A to activate discrete cellular signaling pathways involved in growth control.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cell Proliferation , Mitogen-Activated Protein Kinases/metabolism , Phosphoprotein Phosphatases/metabolism , Signal Transduction/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Blotting, Western , Cell Line , Immunoprecipitation , Mutation/genetics , Phosphorylation , Rats
13.
Mol Cell Biol ; 26(20): 7345-57, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16894031

ABSTRACT

Oncogenic potential is associated with translational regulation, and the prevailing view is that oncogenes use mTOR-dependent pathways to up-regulate the synthesis of proteins critical for transformation. In this study, we show that RalA, a key mediator of Ras transformation, is also linked to the translational machinery. At least part of this linkage, however, is independent of mTOR and acts through RalBP1 to suppress cdc42-mediated activation of S6 kinase and the translation of the antiapoptotic protein FLIP(S). This action, rather than contributing to transformation, opens a latent tumor-suppressive mechanism that can be activated by tumor necrosis factor-related apoptosis-inducing ligand. These results show that the translational machinery is linked to tumor suppression as well as cell-proliferative pathways and that the reestablishment of cell death pathways by activation of the Ral oncogenic program provides a means for selective therapeutic targeting of Ral-driven malignancies.


Subject(s)
Protein Biosynthesis , Protein Kinases/metabolism , ral GTP-Binding Proteins/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Astrocytes/cytology , Astrocytes/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein , Cell Death , Cell Line , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Mice , Phosphorylation , Protein Binding , Protein Kinases/genetics , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , TOR Serine-Threonine Kinases , Tumor Necrosis Factor-alpha/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , ral GTP-Binding Proteins/genetics
14.
Methods Enzymol ; 407: 187-94, 2006.
Article in English | MEDLINE | ID: mdl-16757324

ABSTRACT

Ras family GTPases (RFGs), when in their active GTP-bound state, interact with a wide array of downstream effectors to regulate many biological functions in different cell types. How signal specificity among the closely related family members is achieved is still poorly understood. There is both promiscuity and specificity in the ability of RFGs to interact with and regulate the various effector families, as well as isoforms within those families. RFGs seem to have individual blueprints of effector interactions, and specificity should be considered in the context of the full spectrum of effectors they regulate. The sequencing of the genome has identified a remarkably diverse number of proteins with domains homologous to the Ras-binding domain (RBD) of known Ras effectors and, thus, with the potential to interact with Ras and/or other RFGs. In addition, other proteins without known RBD types are known to behave as RFG effectors, suggesting even more complexity in the number of effector interactions. Determining which of these many candidates are "true" effectors and characterizing their specificity is a critical step to understanding the specific signaling properties and biological functions of the various RFGs.


Subject(s)
ras Proteins/metabolism , Animals , GTP Phosphohydrolases/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Transfection , raf Kinases/metabolism
15.
Mol Cell ; 22(2): 217-30, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16630891

ABSTRACT

Ras family GTPases (RFGs) are known to share many regulatory and effector proteins. How signaling and biological specificity is achieved is poorly understood. Using a proteomics approach, we have identified a complex comprised of Shoc2/Sur-8 and the catalytic subunit of protein phosphatase 1 (PP1c) as a highly specific M-Ras effector. M-Ras targets Shoc2-PP1c to stimulate Raf activity by dephosphorylating the S259 inhibitory site of Raf proteins bound to other molecules of M-Ras or Ras. Therefore, distinct RFGs, through independent effectors, can regulate different steps in the activation of Raf kinases. Shoc2 function is essential for activation of the MAPK pathway by growth factors. Furthermore, in tumor cells with Ras gene mutations, inhibition of Shoc2 expression inhibits MAPK, but not PI3K activity. We propose that the Shoc2-PP1c holoenzyme provides an attractive therapeutic target for inhibition of the MAPK pathway in cancer.


Subject(s)
Fibroblast Growth Factors/chemistry , Phosphoprotein Phosphatases/chemistry , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Repressor Proteins/chemistry , ras Proteins/metabolism , Blotting, Western , Carcinoma/metabolism , Carcinoma/pathology , Catalytic Domain , Cell Line , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Epidermal Growth Factor/pharmacology , Green Fluorescent Proteins/metabolism , HCT116 Cells , HeLa Cells , Holoenzymes/chemistry , Holoenzymes/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mass Spectrometry , Mitogen-Activated Protein Kinases/metabolism , Precipitin Tests , Protein Phosphatase 1 , Protein Structure, Tertiary , Proteomics/methods , RNA Interference , RNA, Small Interfering/metabolism , Retroviridae/genetics , ras Proteins/isolation & purification
16.
Cancer Cell ; 9(4): 243-4, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16616329

ABSTRACT

H-Ras, N-Ras, and K-Ras proteins have distinct biological properties, despite ubiquitous expression and similar affinities for regulators and effectors. C-terminal hypervariable regions that distinguish H-Ras, N-Ras, and K-Ras proteins direct them to distinct membrane compartments, where they may encounter regulators and effectors at different local concentrations. Jura and coworkers now report that these membrane-targeting domains direct differential ubiquitination of Ras proteins and so provide a molecular mechanism to explain the sorting process and, perhaps, some of the dramatic differences in biological potency among H-Ras, N-Ras, and K-Ras proteins.


Subject(s)
Signal Transduction , Ubiquitin/metabolism , ras Proteins/metabolism , Animals , Humans , Protein Binding , ras Proteins/genetics
17.
Science ; 311(5765): 1287-90, 2006 Mar 03.
Article in English | MEDLINE | ID: mdl-16439621

ABSTRACT

Cardio-facio-cutaneous (CFC) syndrome is a sporadic developmental disorder involving characteristic craniofacial features, cardiac defects, ectodermal abnormalities, and developmental delay. We demonstrate that heterogeneous de novo missense mutations in three genes within the mitogen-activated protein kinase (MAPK) pathway cause CFC syndrome. The majority of cases (18 out of 23) are caused by mutations in BRAF, a gene frequently mutated in cancer. Of the 11 mutations identified, two result in amino acid substitutions that occur in tumors, but most are unique and suggest previously unknown mechanisms of B-Raf activation. Furthermore, three of five individuals without BRAF mutations had missense mutations in either MEK1 or MEK2, downstream effectors of B-Raf. Our findings highlight the involvement of the MAPK pathway in human development and will provide a molecular diagnosis of CFC syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Germ-Line Mutation , Mitogen-Activated Protein Kinases/metabolism , Adolescent , Adult , Amino Acid Substitution , Child , Child, Preschool , Craniofacial Abnormalities/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Growth Disorders/genetics , Heart Defects, Congenital/genetics , Humans , Infant , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Male , Mitogen-Activated Protein Kinases/genetics , Mutation, Missense , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , Skin Abnormalities/genetics , Syndrome , Transfection
18.
Cancer Cell ; 8(2): 111-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16098464

ABSTRACT

The EphA2 receptor tyrosine kinase is frequently overexpressed in many cancers, including 40% of breast cancers. Here, we show that EphA2 is a direct transcriptional target of the Ras-Raf-MAPK pathway and that ligand-stimulated EphA2 attenuates the growth factor-induced activation of Ras. Thus, a negative feedback loop is created that regulates Ras activity. Interestingly, the expression of EphA2 and ephrin-A1 is mutually exclusive in a panel of 28 breast cancer cell lines. We show that the MAPK pathway inhibits ephrin-A1 expression, and the ligand expression inhibits EphA2 levels contributing to the receptor-ligand reciprocal expression pattern in these cell lines. Our results suggest that an escape from the negative effects of this interaction may be important in the development of cancer.


Subject(s)
Breast Neoplasms/metabolism , Down-Regulation , Receptor, EphA2/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , Ephrin-A1/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , NIH 3T3 Cells , Oncogene Proteins v-erbB/metabolism , Receptor, EphA2/genetics , Signal Transduction , Transcription, Genetic , raf Kinases/metabolism
19.
Cancer Cell ; 7(3): 205-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15766656

ABSTRACT

Ras proteins send signals through multiple effector pathways. The Raf/MEK/MAPK and PI 3' kinase pathways are well-validated Ras effectors in human cancers, but many other candidate pathways could be equally important. RalGDS is such a candidate: in a new paper from Chris Marshall's group, an important role for RalGDS in Ras transformation in vivo has been established for the first time. Mice lacking RalGDS are defective in tumor formation, possibly because of increased apoptosis in Ras-driven tumors. The hunt for a clear role for RalGDS activation in human cancer is on.


Subject(s)
Signal Transduction/physiology , ral Guanine Nucleotide Exchange Factor/metabolism , ras Proteins/metabolism , Animals , Humans , Mice , Neoplasms/metabolism
20.
Mol Cell Biol ; 24(11): 4943-54, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15143186

ABSTRACT

Ras family GTPases (RFGs) regulate signaling pathways that control multiple biological processes. How signaling specificity among the closely related family members is achieved is poorly understood. We have taken a proteomics approach to signaling by RFGs, and we have analyzed interactions of a panel of RFGs with a comprehensive group of known and potential effectors. We have found remarkable differences in the ability of RFGs to regulate the various isoforms of known effector families. We have also identified several proteins as novel effectors of RFGs with differential binding specificities to the various RFGs. We propose that specificity among RFGs is achieved by the differential regulation of combinations of effector families as well as by the selective regulation of different isoforms within an effector family. An understanding of this new level of complexity in the signaling pathways regulated by RFGs is necessary to understand how they carry out their many cellular functions. It will also likely have critical implications in the treatment of human diseases such as cancer.


Subject(s)
GTP Phosphohydrolases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms , Protein Structure, Tertiary , Substrate Specificity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...