Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
1.
Cell Rep ; 42(7): 112670, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37392382

ABSTRACT

Eph receptors and their ephrin ligands are viewed as promising targets for cancer treatment; however, targeting them is hindered by their context-dependent functionalities. To circumvent this, we explore molecular landscapes underlying their pro- and anti-malignant activities. Using unbiased bioinformatics approaches, we construct a cancer-related network of genetic interactions (GIs) of all Ephs and ephrins to assist in their therapeutic manipulation. We also apply genetic screening and BioID proteomics and integrate them with machine learning approaches to select the most relevant GIs of one Eph receptor, EPHB6. This identifies a crosstalk between EPHB6 and EGFR, and further experiments confirm the ability of EPHB6 to modulate EGFR signaling, enhancing the proliferation of cancer cells and tumor development. Taken together, our observations show EPHB6 involvement in EGFR action, suggesting its targeting might be beneficial in EGFR-dependent tumors, and confirm that the Eph family genetic interactome presented here can be effectively exploited in developing cancer treatment approaches.


Subject(s)
Ephrins , Neoplasms , Ephrins/genetics , Proteomics , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Signal Transduction , ErbB Receptors/genetics , Neoplasms/genetics
2.
Clin Cancer Res ; 29(14): 2686-2701, 2023 07 14.
Article in English | MEDLINE | ID: mdl-36976175

ABSTRACT

PURPOSE: Accumulating analyses of pro-oncogenic molecular mechanisms triggered a rapid development of targeted cancer therapies. Although many of these treatments produce impressive initial responses, eventual resistance onset is practically unavoidable. One of the main approaches for preventing this refractory condition relies on the implementation of combination therapies. This includes dual-specificity reagents that affect both of their targets with a high level of selectivity. Unfortunately, selection of target combinations for these treatments is often confounded by limitations in our understanding of tumor biology. Here, we describe and validate a multipronged unbiased strategy for predicting optimal co-targets for bispecific therapeutics. EXPERIMENTAL DESIGN: Our strategy integrates ex vivo genome-wide loss-of-function screening, BioID interactome profiling, and gene expression analysis of patient data to identify the best fit co-targets. Final validation of selected target combinations is done in tumorsphere cultures and xenograft models. RESULTS: Integration of our experimental approaches unambiguously pointed toward EGFR and EPHA2 tyrosine kinase receptors as molecules of choice for co-targeting in multiple tumor types. Following this lead, we generated a human bispecific anti-EGFR/EPHA2 antibody that, as predicted, very effectively suppresses tumor growth compared with its prototype anti-EGFR therapeutic antibody, cetuximab. CONCLUSIONS: Our work not only presents a new bispecific antibody with a high potential for being developed into clinically relevant biologics, but more importantly, successfully validates a novel unbiased strategy for selecting biologically optimal target combinations. This is of a significant translational relevance, as such multifaceted unbiased approaches are likely to augment the development of effective combination therapies for cancer treatment. See related commentary by Kumar, p. 2570.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , ErbB Receptors/metabolism , Cell Line, Tumor , Cetuximab/pharmacology , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/immunology , Neoplasms/drug therapy , Neoplasms/genetics
3.
Cell Rep ; 40(1): 111031, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35793621

ABSTRACT

EPH receptors (EPHRs) constitute the largest family among receptor tyrosine kinases in humans. They are mainly involved in short-range cell-cell communication events that regulate cell adhesion, migration, and boundary formation. However, the molecular mechanisms by which EPHRs control these processes are less understood. To address this, we unravel EPHR-associated complexes under native conditions using mass-spectrometry-based BioID proximity labeling. We obtain a composite proximity network from EPHA4, -B2, -B3, and -B4 that comprises 395 proteins, most of which were not previously linked to EPHRs. We examine the contribution of several BioID-identified candidates via loss-of-function in an EPHR-dependent cell-segregation assay. We find that the signaling scaffold PAR-3 is required for cell sorting and that EPHRs directly phosphorylate PAR-3. We also delineate a signaling complex involving the C-terminal SRC kinase (CSK), whose recruitment to PAR-3 is dependent on EPHR signals. Our work describes signaling networks by which EPHRs regulate cellular phenotypes.


Subject(s)
Receptors, Eph Family , Signal Transduction , CSK Tyrosine-Protein Kinase , Cell Communication , Software
4.
J Am Soc Nephrol ; 33(8): 1546-1567, 2022 08.
Article in English | MEDLINE | ID: mdl-35906089

ABSTRACT

BACKGROUND: Maintenance of the kidney filtration barrier requires coordinated interactions between podocytes and the underlying glomerular basement membrane (GBM). GBM ligands bind podocyte integrins, which triggers actin-based signaling events critical for adhesion. Nck1/2 adaptors have emerged as essential regulators of podocyte cytoskeletal dynamics. However, the precise signaling mechanisms mediated by Nck1/2 adaptors in podocytes remain to be fully elucidated. METHODS: We generated podocytes deficient in Nck1 and Nck2 and used transcriptomic approaches to profile expression differences. Proteomic techniques identified specific binding partners for Nck1 and Nck2 in podocytes. We used cultured podocytes and mice deficient in Nck1 and/or Nck2, along with podocyte injury models, to comprehensively verify our findings. RESULTS: Compound loss of Nck1/2 altered expression of genes involved in actin binding, cell adhesion, and extracellular matrix composition. Accordingly, Nck1/2-deficient podocytes showed defects in actin organization and cell adhesion in vitro, with podocyte detachment and altered GBM morphology present in vivo. We identified distinct interactomes for Nck1 and Nck2 and uncovered a mechanism by which Nck1 and Nck2 cooperate to regulate actin bundling at focal adhesions via α actinin-4. Furthermore, loss of Nck1 or Nck2 resulted in increased matrix deposition in vivo, with more prominent defects in Nck2-deficient mice, consistent with enhanced susceptibility to podocyte injury. CONCLUSION: These findings reveal distinct, yet complementary, roles for Nck proteins in regulating podocyte adhesion, controlling GBM composition, and sustaining filtration barrier integrity.


Subject(s)
Podocytes , Actinin/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Glomerular Basement Membrane/metabolism , Mice , Oncogene Proteins/metabolism , Podocytes/metabolism , Proteomics
5.
Trends Biochem Sci ; 47(9): 772-784, 2022 09.
Article in English | MEDLINE | ID: mdl-35562294

ABSTRACT

The assembly of complexes following the detection of extracellular signals is often controlled by signaling proteins comprising multiple peptide binding modules. The SRC homology (SH)3 family represents the archetypical modular protein interaction module, with ~300 annotated SH3 domains in humans that regulate an impressive array of signaling processes. We review recent findings regarding the allosteric contributions of SH3 domains host protein context, their phosphoregulation, and their roles in phase separation that challenge the simple model in which SH3s are considered to be portable domains binding to specific proline-rich peptide motifs.


Subject(s)
Proteins , src Homology Domains , Binding Sites , Humans , Peptides/metabolism , Protein Binding , Proteins/metabolism
6.
J Biol Chem ; 297(5): 101291, 2021 11.
Article in English | MEDLINE | ID: mdl-34634301

ABSTRACT

Metabolic dysfunction is a major driver of tumorigenesis. The serine/threonine kinase mechanistic target of rapamycin (mTOR) constitutes a key central regulator of metabolic pathways promoting cancer cell proliferation and survival. mTOR activity is regulated by metabolic sensors as well as by numerous factors comprising the phosphatase and tensin homolog/PI3K/AKT canonical pathway, which are often mutated in cancer. However, some cancers displaying constitutively active mTOR do not carry alterations within this canonical pathway, suggesting alternative modes of mTOR regulation. Since DEPTOR, an endogenous inhibitor of mTOR, was previously found to modulate both mTOR complexes 1 and 2, we investigated the different post-translational modification that could affect its inhibitory function. We found that tyrosine (Tyr) 289 phosphorylation of DEPTOR impairs its interaction with mTOR, leading to increased mTOR activation. Using proximity biotinylation assays, we identified SYK (spleen tyrosine kinase) as a kinase involved in DEPTOR Tyr 289 phosphorylation in an ephrin (erythropoietin-producing hepatocellular carcinoma) receptor-dependent manner. Altogether, our work reveals that phosphorylation of Tyr 289 of DEPTOR represents a novel molecular switch involved in the regulation of both mTOR complex 1 and mTOR complex 2.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Protein Processing, Post-Translational , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Phosphorylation , TOR Serine-Threonine Kinases/genetics , Tyrosine/genetics , Tyrosine/metabolism
7.
Nat Commun ; 12(1): 1597, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712617

ABSTRACT

Protein-protein interactions (PPIs) between modular binding domains and their target peptide motifs are thought to largely depend on the intrinsic binding specificities of the domains. The large family of SRC Homology 3 (SH3) domains contribute to cellular processes via their ability to support such PPIs. While the intrinsic binding specificities of SH3 domains have been studied in vitro, whether each domain is necessary and sufficient to define PPI specificity in vivo is largely unknown. Here, by combining deletion, mutation, swapping and shuffling of SH3 domains and measurements of their impact on protein interactions in yeast, we find that most SH3s do not dictate PPI specificity independently from their host protein in vivo. We show that the identity of the host protein and the position of the SH3 domains within their host are critical for PPI specificity, for cellular functions and for key biophysical processes such as phase separation. Our work demonstrates the importance of the interplay between a modular PPI domain such as SH3 and its host protein in establishing specificity to wire PPI networks. These findings will aid understanding how protein networks are rewired during evolution and in the context of mutation-driven diseases such as cancer.


Subject(s)
Protein Interaction Maps , Proteins/chemistry , src Homology Domains , HEK293 Cells , Humans , Protein Interaction Domains and Motifs , Proteins/metabolism , Saccharomyces cerevisiae/metabolism , src Homology Domains/genetics
8.
Mol Cell Proteomics ; 20: 100064, 2021.
Article in English | MEDLINE | ID: mdl-33640491

ABSTRACT

Prostate cancer (PCa) is the most frequently diagnosed cancer in men and the third cause of cancer mortality. PCa initiation and growth are driven by the androgen receptor (AR). The AR is activated by androgens such as testosterone and controls prostatic cell proliferation and survival. Here, we report an AR signaling network generated using BioID proximity labeling proteomics in androgen-dependent LAPC4 cells. We identified 31 AR-associated proteins in nonstimulated cells. Strikingly, the AR signaling network increased to 182 and 200 proteins, upon 24 h or 72 h of androgenic stimulation, respectively, for a total of 267 nonredundant AR-associated candidates. Among the latter group, we identified 213 proteins that were not previously reported in databases. Many of these new AR-associated proteins are involved in DNA metabolism, RNA processing, and RNA polymerase II transcription. Moreover, we identified 44 transcription factors, including the Kru¨ppel-like factor 4 (KLF4), which were found interacting in androgen-stimulated cells. Interestingly, KLF4 repressed the well-characterized AR-dependent transcription of the KLK3 (PSA) gene; AR and KLF4 also colocalized genome-wide. Taken together, our data report an expanded high-confidence proximity network for AR, which will be instrumental to further dissect the molecular mechanisms underlying androgen signaling in PCa cells.


Subject(s)
Receptors, Androgen/metabolism , Cell Line , Humans , Kallikreins/genetics , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Prostate-Specific Antigen/genetics , Receptors, Androgen/genetics
9.
J Biol Chem ; 295(31): 10535-10559, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32299913

ABSTRACT

SHC adaptor protein (SHCA) and lipoma-preferred partner (LPP) mediate transforming growth factor ß (TGFß)-induced breast cancer cell migration and invasion. Reduced expression of either protein diminishes breast cancer lung metastasis, but the reason for this effect is unclear. Here, using total internal reflection fluorescence (TIRF) microscopy, we found that TGFß enhanced the assembly and disassembly rates of paxillin-containing adhesions in an SHCA-dependent manner through the phosphorylation of the specific SHCA tyrosine residues Tyr-239, Tyr-240, and Tyr-313. Using a BioID proximity labeling approach, we show that SHCA exists in a complex with a variety of actin cytoskeletal proteins, including paxillin and LPP. Consistent with a functional interaction between SHCA and LPP, TGFß-induced LPP localization to cellular adhesions depended on SHCA. Once localized to the adhesions, LPP was required for TGFß-induced increases in cell migration and adhesion dynamics. Mutations that impaired LPP localization to adhesions (mLIM1) or impeded interactions with the actin cytoskeleton via α-actinin (ΔABD) abrogated migratory responses to TGFß. Live-cell TIRF microscopy revealed that SHCA clustering at the cell membrane preceded LPP recruitment. We therefore hypothesize that, in the presence of TGFß, SHCA promotes the formation of small, dynamic adhesions by acting as a nucleator of focal complex formation. Finally, we defined a previously unknown function for SHCA in the formation of invadopodia, a process that also required LPP. Our results reveal that SHCA controls the formation and function of adhesions and invadopodia, two key cellular structures required for breast cancer metastasis.


Subject(s)
Cell Movement , Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Podosomes/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Cell Adhesion , Cell Line, Transformed , Cytoskeletal Proteins/genetics , Female , LIM Domain Proteins/genetics , Mice , Paxillin/genetics , Paxillin/metabolism , Podosomes/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Transforming Growth Factor beta
10.
Cell Chem Biol ; 27(6): 657-667.e6, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32220335

ABSTRACT

Targeting protein-protein interactions (PPIs) is a promising approach in the development of drugs for many indications. 14-3-3 proteins are a family of phosphoprotein-binding molecules with critical functions in dozens of cell signaling networks. 14-3-3s are abundant in the central nervous system, and the small molecule fusicoccin-A (FC-A), a tool compound that can be used to manipulate 14-3-3 PPIs, enhances neurite outgrowth in cultured neurons. New semisynthetic FC-A derivatives with improved binding affinity for 14-3-3 complexes have recently been developed. Here, we use a series of screens that identify these compounds as potent inducers of neurite outgrowth through a polypharmacological mechanism. Using proteomics and X-ray crystallography, we discover that these compounds extensively regulate the 14-3-3 interactome by stabilizing specific PPIs, while disrupting others. These results provide new insights into the development of drugs to target 14-3-3 PPIs, a potential therapeutic strategy for CNS diseases.


Subject(s)
14-3-3 Proteins/antagonists & inhibitors , Glycosides/pharmacology , Neurites/drug effects , Small Molecule Libraries/pharmacology , 14-3-3 Proteins/isolation & purification , 14-3-3 Proteins/metabolism , Animals , Cells, Cultured , Crystallography, X-Ray , Dose-Response Relationship, Drug , Female , Glycosides/chemistry , Male , Models, Molecular , Molecular Conformation , Neurites/metabolism , Neuronal Outgrowth/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/chemistry
11.
Mol Cell Proteomics ; 17(10): 1979-1990, 2018 10.
Article in English | MEDLINE | ID: mdl-30002203

ABSTRACT

Signals from cell surface receptors are often relayed via adaptor proteins. NCK1 and NCK2 are Src-Homology (SH) 2 and 3 domain adaptors that regulate processes requiring a remodeling of the actin cytoskeleton. Evidence from gene inactivation in mouse suggests that NCK1 and NCK2 are functionally redundant, although recent reports support the idea of unique functions for NCK1 and NCK2. We sought to examine this question further by delineating NCK1- and NCK2-specific signaling networks. We used both affinity purification-mass spectrometry and BioID proximity labeling to identify NCK1/2 signaling networks comprised of 98 proteins. Strikingly, we found 30 proteins restricted to NCK1 and 28 proteins specifically associated with NCK2, suggesting differences in their function. We report that Nck2-/-, but not Nck1-/- mouse embryo fibroblasts (MEFs) are multinucleated and display extended protrusions reminiscent of intercellular bridges, which correlate with an extended time spent in cytokinesis as well as a failure of a significant proportion of cells to complete abscission. Our data also show that the midbody of NCK2-deficient cells is not only increased in length, but also altered in composition, as judged by the mislocalization of AURKB, PLK1 and ECT2. Finally, we show that NCK2 function during cytokinesis requires its SH2 domain. Taken together, our data delineate the first high-confidence interactome for NCK1/2 adaptors and highlight several proteins specifically associated with either protein. Thus, contrary to what is generally accepted, we demonstrate that NCK1 and NCK2 are not completely redundant, and shed light on a previously uncharacterized function for the NCK2 adaptor protein in cell division.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytokinesis , Oncogene Proteins/metabolism , Proteomics , Adaptor Proteins, Signal Transducing/chemistry , Animals , HEK293 Cells , HeLa Cells , Humans , Mass Spectrometry , Mice , Oncogene Proteins/chemistry , Protein Interaction Mapping , Protein Transport , Structure-Activity Relationship , src Homology Domains
12.
Mol Cell ; 70(6): 995-1007.e11, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29910111

ABSTRACT

Phosphotyrosine (pTyr) signaling has evolved into a key cell-to-cell communication system. Activated receptor tyrosine kinases (RTKs) initiate several pTyr-dependent signaling networks by creating the docking sites required for the assembly of protein complexes. However, the mechanisms leading to network disassembly and its consequence on signal transduction remain essentially unknown. We show that activated RTKs terminate downstream signaling via the direct phosphorylation of an evolutionarily conserved Tyr present in most SRC homology (SH) 3 domains, which are often part of key hub proteins for RTK-dependent signaling. We demonstrate that the direct EPHA4 RTK phosphorylation of adaptor protein NCK SH3s at these sites results in the collapse of signaling networks and abrogates their function. We also reveal that this negative regulation mechanism is shared by other RTKs. Our findings uncover a conserved mechanism through which RTKs rapidly and reversibly terminate downstream signaling while remaining in a catalytically active state on the plasma membrane.


Subject(s)
Receptor Protein-Tyrosine Kinases/physiology , Receptor, EphA4/metabolism , src Homology Domains/physiology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Cell Communication , Drosophila/metabolism , HEK293 Cells , HeLa Cells , Humans , Ligands , Oncogene Proteins/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Tyrosine/metabolism
13.
Dis Model Mech ; 11(3)2018 03 13.
Article in English | MEDLINE | ID: mdl-29590634

ABSTRACT

The RAS/MAPK signaling pathway is one of the most investigated pathways, owing to its established role in numerous cellular processes and implication in cancer. Germline mutations in genes encoding members of the RAS/MAPK pathway also cause severe developmental syndromes collectively known as RASopathies. These syndromes share overlapping characteristics, including craniofacial dysmorphology, cardiac malformations, cutaneous abnormalities and developmental delay. Cardio-facio-cutaneous syndrome (CFC) is a rare RASopathy associated with mutations in BRAF, KRAS, MEK1 (MAP2K1) and MEK2 (MAP2K2). MEK1 and MEK2 mutations are found in ∼25% of the CFC patients and the MEK1Y130C substitution is the most common one. However, little is known about the origins and mechanisms responsible for the development of CFC. To our knowledge, no mouse model carrying RASopathy-linked Mek1 or Mek2 gene mutations has been reported. To investigate the molecular and developmental consequences of the Mek1Y130C mutation, we generated a mouse line carrying this mutation. Analysis of mice from a Mek1 allelic series revealed that the Mek1Y130C allele expresses both wild-type and Y130C mutant forms of MEK1. However, despite reduced levels of MEK1 protein and the lower abundance of MEK1 Y130C protein than wild type, Mek1Y130C mutants showed increased ERK (MAPK) protein activation in response to growth factors, supporting a role for MEK1 Y130C in hyperactivation of the RAS/MAPK pathway, leading to CFC. Mek1Y130C mutant mice exhibited pulmonary artery stenosis, cranial dysmorphia and neurological anomalies, including increased numbers of GFAP+ astrocytes and Olig2+ oligodendrocytes in regions of the cerebral cortex. These data indicate that the Mek1Y130C mutation recapitulates major aspects of CFC, providing a new animal model to investigate the physiopathology of this RASopathy. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Ectodermal Dysplasia/genetics , Failure to Thrive/genetics , Heart Defects, Congenital/genetics , MAP Kinase Kinase 1/genetics , Mutation/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Brain/pathology , Cell Count , Embryo, Mammalian/cytology , Facies , Fibroblasts/enzymology , Gene Duplication , Glial Fibrillary Acidic Protein/metabolism , Humans , MAP Kinase Kinase 1/chemistry , MAP Kinase Signaling System/genetics , Mice , Mice, Mutant Strains , Oligodendrocyte Transcription Factor 2/metabolism
14.
J Proteomics ; 189: 39-47, 2018 10 30.
Article in English | MEDLINE | ID: mdl-29425736

ABSTRACT

Protein phosphorylation often regulates interactions between components of signalling networks. Its tight regulation by opposing actions of kinases and phosphatases contribute to making this modification key in controlling the dynamic architecture of phosphorylation-dependent networks. The introduction in cell signalling research of long-standing targeted proteomics approaches such as selected reaction monitoring (SRM) combined with the development of state-of-the-art methods such as parallel reaction monitoring (PRM) and data-independent analysis (DIA), have allowed delineating temporal quantitative profiles of interactions networks. This review summarizes how targeted proteomics analyses have recently contributed to our comprehension of phosphorylation-dependent protein interaction networks and proposes how these could be applied to address clinical problems. BIOLOGICAL SIGNIFICANCE: Intracellular communication and information processing are performed by context-specific protein interaction networks that are often regulated by protein phosphorylation. Quantification of dynamic changes within these signalling networks is of critical importance to understand cell biology in normal and disease states, but remains challenging. Targeted proteomics approaches have contributed greatly to our understanding of these phosphorylation-dependent signalling networks.


Subject(s)
Phosphotransferases/metabolism , Proteome/metabolism , Proteomics/methods , Humans , Metabolic Networks and Pathways/physiology , Phosphorylation , Protein Processing, Post-Translational/physiology , Proteome/analysis
15.
Sci Rep ; 7(1): 11514, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912526

ABSTRACT

HER2/ErbB2 is overexpressed in a significant fraction of breast tumours and is associated with a poor prognosis. The adaptor protein GRB2 interacts directly with activated HER2 and is sufficient to transmit oncogenic signals. However, the consequence of HER2 activation on global GRB2 signalling networks is poorly characterized. We performed GRB2 affinity purification combined with mass spectrometry analysis of associated proteins in a HER2+ breast cancer model to delineate GRB2-nucleated protein interaction networks. We report the identification of the transmembrane protein MPZL1 as a new GRB2-associated protein. Our data show that the PTPN11 tyrosine phosphatase acts as a scaffold to bridge the association between GRB2 and MPZL1 in a phosphotyrosine-dependent manner. We further demonstrate that the formation of this MPZL1-PTPN11-GRB2 complex is triggered by cell attachment to fibronectin. Thus, our data support the importance of this new signalling complex in the control of cell adhesion of HER2+ breast cancer cells, a key feature of the metastatic process.


Subject(s)
Breast Neoplasms/pathology , GRB2 Adaptor Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Protein Multimerization , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Cell Adhesion , Cell Line , Chromatography, Affinity , Female , Fibronectins/metabolism , GRB2 Adaptor Protein/isolation & purification , Humans , Mass Spectrometry , Protein Binding , Protein Interaction Mapping
16.
Neuron ; 93(5): 1082-1093.e5, 2017 Mar 08.
Article in English | MEDLINE | ID: mdl-28279353

ABSTRACT

Damaged central nervous system (CNS) neurons have a poor ability to spontaneously regenerate, causing persistent functional deficits after injury. Therapies that stimulate axon growth are needed to repair CNS damage. 14-3-3 adaptors are hub proteins that are attractive targets to manipulate cell signaling. We identify a positive role for 14-3-3s in axon growth and uncover a developmental regulation of the phosphorylation and function of 14-3-3s. We show that fusicoccin-A (FC-A), a small-molecule stabilizer of 14-3-3 protein-protein interactions, stimulates axon growth in vitro and regeneration in vivo. We show that FC-A stabilizes a complex between 14-3-3 and the stress response regulator GCN1, inducing GCN1 turnover and neurite outgrowth. These findings show that 14-3-3 adaptor protein complexes are druggable targets and identify a new class of small molecules that may be further optimized for the repair of CNS damage.


Subject(s)
14-3-3 Proteins/metabolism , Axons/metabolism , Glycosides/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Cells, Cultured , Mice , Nerve Regeneration/physiology , Rats, Sprague-Dawley
17.
J Biol Chem ; 292(14): 5748-5759, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28213521

ABSTRACT

Proteins of the Src homology and collagen (Shc) family are typically involved in signal transduction events involving Ras/MAPK and PI3K/Akt pathways. In the nervous system, they function proximal to the neurotrophic factors that regulate cell survival, differentiation, and neuron-specific characteristics. The least characterized homolog, ShcD, is robustly expressed in the developing and mature nervous system, but its contributions to neural cell circuitry are largely uncharted. We now report that ShcD binds to active Ret, TrkA, and TrkB neurotrophic factor receptors predominantly via its phosphotyrosine-binding (PTB) domain. However, in contrast to the conventional Shc adaptors, ShcD suppresses distal phosphorylation of the Erk MAPK. Accordingly, genetic knock-out of mouse ShcD enhances Erk phosphorylation in the brain. In cultured cells, this capacity is tightly aligned to phosphorylation of ShcD CH1 region tyrosine motifs, which serve as docking platforms for signal transducers, such as Grb2. Erk suppression is relieved through independent mutagenesis of the PTB domain and the CH1 tyrosine residues, and successive substitution of these tyrosines breaks the interaction between ShcD and Grb2, thereby promoting TrkB-Grb2 association. Erk phosphorylation can also be restored in the presence of wild type ShcD through Grb2 overexpression. Conversely, mutation of the ShcD SH2 domain results in enhanced repression of Erk. Although the SH2 domain is a less common binding interface in Shc proteins, we demonstrate that it associates with the Ptpn11 (Shp2) phosphatase, which in turn regulates ShcD tyrosine phosphorylation. We therefore propose a model whereby ShcD competes with neurotrophic receptors for Grb2 binding and opposes activation of the MAPK cascade.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Receptor, trkA/metabolism , Shc Signaling Adaptor Proteins/metabolism , Amino Acid Motifs , Cell Line , Extracellular Signal-Regulated MAP Kinases/genetics , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Membrane Glycoproteins/genetics , Phosphorylation/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-ret/genetics , Receptor, trkA/genetics , Receptor, trkB , Shc Signaling Adaptor Proteins/genetics
18.
Methods Mol Biol ; 1458: 339-47, 2016.
Article in English | MEDLINE | ID: mdl-27581032

ABSTRACT

A precisely controlled network of protein-protein interactions constitutes the basis for functional signaling pathways. This equilibrium is more often than not disrupted in cancer cells, by the aberrant expression or activation of oncogenic proteins. Therefore, the analysis of protein interaction networks in cancer cells has become crucial to expand our comprehension of the molecular underpinnings of tumor formation and progression. This protocol describes a sample preparation method for the analysis of signaling complexes by mass spectrometry (MS), following the affinity purification of a protein of interest from a cancer cell line or a solid tumor. In particular, we provide a spin tip-based protease digestion procedure that offers a more rapid and controlled alternative to other gel-based and gel-free methods. This sample preparation protocol represents a useful strategy to identify protein interactions and to gain insight into the molecular mechanisms that contribute to a given cancer phenotype.


Subject(s)
Mass Spectrometry , Neoplasms/metabolism , Protein Interaction Mapping , Protein Interaction Maps , Cell Line, Tumor , Humans , Mass Spectrometry/methods , Protein Binding , Protein Interaction Mapping/methods
19.
Proc Natl Acad Sci U S A ; 112(14): 4501-6, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25831502

ABSTRACT

Cellular processes and homeostasis control in eukaryotic cells is achieved by the action of regulatory proteins such as protein kinase A (PKA). Although the outbound signals from PKA directed to processes such as metabolism, growth, and aging have been well charted, what regulates this conserved regulator remains to be systematically identified to understand how it coordinates biological processes. Using a yeast PKA reporter assay, we identified genes that influence PKA activity by measuring protein-protein interactions between the regulatory and the two catalytic subunits of the PKA complex in 3,726 yeast genetic-deletion backgrounds grown on two carbon sources. Overall, nearly 500 genes were found to be connected directly or indirectly to PKA regulation, including 80 core regulators, denoting a wide diversity of signals regulating PKA, within and beyond the described upstream linear pathways. PKA regulators span multiple processes, including the antagonistic autophagy and methionine biosynthesis pathways. Our results converge toward mechanisms of PKA posttranslational regulation by lysine acetylation, which is conserved between yeast and humans and that, we show, regulates protein complex formation in mammals and carbohydrate storage and aging in yeast. Taken together, these results show that the extent of PKA input matches with its output, because this kinase receives information from upstream and downstream processes, and highlight how biological processes are interconnected and coordinated by PKA.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Signal Transduction , Acetylation , Amino Acid Sequence , Animals , Autophagy , Cyclic AMP/metabolism , Galactose/chemistry , Glucose/chemistry , HEK293 Cells , Homeostasis , Humans , Luciferases, Renilla/metabolism , Methionine/chemistry , Molecular Sequence Data , Phylogeny , Protein Processing, Post-Translational , Rats , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , TOR Serine-Threonine Kinases/metabolism
20.
J Cell Biol ; 204(4): 487-95, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24515345

ABSTRACT

During epithelial cell polarization, Yurt (Yrt) is initially confined to the lateral membrane and supports the stability of this membrane domain by repressing the Crumbs-containing apical machinery. At late stages of embryogenesis, the apical recruitment of Yrt restricts the size of the apical membrane. However, the molecular basis sustaining the spatiotemporal dynamics of Yrt remains undefined. In this paper, we report that atypical protein kinase C (aPKC) phosphorylates Yrt to prevent its premature apical localization. A nonphosphorylatable version of Yrt dominantly dismantles the apical domain, showing that its aPKC-mediated exclusion is crucial for epithelial cell polarity. In return, Yrt counteracts aPKC functions to prevent apicalization of the plasma membrane. The ability of Yrt to bind and restrain aPKC signaling is central for its role in polarity, as removal of the aPKC binding site neutralizes Yrt activity. Thus, Yrt and aPKC are involved in a reciprocal antagonistic regulatory loop that contributes to segregation of distinct and mutually exclusive membrane domains in epithelial cells.


Subject(s)
Cell Polarity , Drosophila Proteins/antagonists & inhibitors , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/metabolism , Epithelial Cells/metabolism , Protein Kinase C/antagonists & inhibitors , Amino Acid Sequence , Animals , Animals, Genetically Modified , Blotting, Western , Cells, Cultured , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Embryo, Nonmammalian/cytology , Epithelial Cells/cytology , Fluorescent Antibody Technique , Immunoenzyme Techniques , Immunoprecipitation , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL