Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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
2.
Cell Cycle ; 19(22): 2963-2981, 2020 11.
Article in English | MEDLINE | ID: mdl-33103553

ABSTRACT

The human adenovirus (Ad) type 2/5 early region 4 (E4) ORF4 protein (E4orf4) exerts a remarkable tumor cell-selective killing activity in mammalian cells. This indicates that E4orf4 can target tumor cell-defining features and is a unique tool to probe cancer cell vulnerabilities. Recently, we found that E4orf4, through an interaction with the polarity protein PAR3, subverts nuclear envelope (NE) remodeling processes in a tumor cell-selective manner. In this Perspective, we outline mechanical signals that modify nuclear dynamics and tumor cell behavior to highlight potential mechanisms for E4orf4's tumoricidal activity. Through an analysis of E4orf4's cellular targets, we define a protein subnetwork that comprises phosphatase systems interconnected to polarity protein hubs, which could contribute to enhanced NE plasticity. We infer that elucidating E4orf4's protein network at a functional level could uncover key mechanisms of NE remodeling that define the tumor cell phenotype.


Subject(s)
Carcinogenesis/metabolism , Neoplasms/metabolism , Nuclear Envelope/metabolism , Stress, Mechanical , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Drosophila/metabolism , Hippo Signaling Pathway/metabolism , Humans , Neoplasms/pathology , Phenotype , Protein Interaction Maps , Protein Phosphatase 2/metabolism
3.
J Cell Biol ; 219(4)2020 04 06.
Article in English | MEDLINE | ID: mdl-32328642

ABSTRACT

The tumor cell-selective killing activity of the adenovirus type 2 early region 4 ORF4 (E4orf4) protein is poorly defined at the molecular level. Here, we show that the tumoricidal effect of E4orf4 is typified by changes in nuclear dynamics that depend on its interaction with the polarity protein Par3 and actomyosin contractility. Mechanistically, E4orf4 induced a high incidence of nuclear bleb formation and repetitive nuclear ruptures, which promoted nuclear efflux of E4orf4 and loss of nuclear integrity. This process was regulated by nucleocytoskeletal connections, Par3 clustering proximal to nuclear lamina folds, and retrograde movement of actin bundles that correlated with nuclear ruptures. Significantly, Par3 also regulated the incidence of spontaneous nuclear ruptures facilitated by the downmodulation of lamins. This work uncovered a novel role for Par3 in controlling the actin-dependent forces acting on the nuclear envelope to remodel nuclear shape, which might be a defining feature of tumor cells that is harnessed by E4orf4.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Viral Proteins/metabolism , Cell Death , HEK293 Cells , HeLa Cells , Humans
4.
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
5.
J Cell Biol ; 217(11): 3853-3862, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30082297

ABSTRACT

Drosophila melanogaster Yurt (Yrt) and its mammalian orthologue EPB41L5 limit apical membrane growth in polarized epithelia. EPB41L5 also supports epithelial-mesenchymal transition and metastasis. Yrt and EPB41L5 contain a four-point-one, ezrin, radixin, and moesin (FERM) domain and a FERM-adjacent (FA) domain. The former contributes to the quaternary structure of 50 human proteins, whereas the latter defines a subfamily of 14 human FERM proteins and fulfills unknown roles. In this study, we show that both Yrt and EPB41L5 oligomerize. Our data also establish that the FERM-FA unit forms an oligomeric interface and that multimerization of Yrt is crucial for its function in epithelial cell polarity regulation. Finally, we demonstrate that aPKC destabilizes the Yrt oligomer to repress its functions, thereby revealing a mechanism through which this kinase supports apical domain formation. Overall, our study highlights a conserved biochemical property of fly and human Yrt proteins, describes a novel function of the FA domain, and further characterizes the molecular mechanisms sustaining epithelial cell polarity.


Subject(s)
Cell Polarity , Drosophila Proteins/metabolism , Epithelial Cells/metabolism , Protein Multimerization , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster , Epithelial Cells/chemistry , Epithelial Cells/cytology , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Domains
6.
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
7.
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
8.
Biochem Cell Biol ; : 1-8, 2018 Apr 30.
Article in English | MEDLINE | ID: mdl-29707960

ABSTRACT

Metal-responsive transcription factor-1 (MTF-1) is a metal-regulatory transcription factor essential for induction of the genes encoding metallothioneins (MTs) in response to transition metal ions. Activation of MTF-1 is dependent on the interaction of zinc with the zinc fingers of the protein. In addition, phosphorylation is essential for MTF-1 transactivation. We previously showed that inhibition of phosphoinositide 3-kinase (PI3K) abrogated Mt expression and metal-induced MTF-1 activation in human hepatocellular carcinoma (HCC) HepG2 and mouse L cells, thus showing that the PI3K signaling pathway positively regulates MTF-1 activity and Mt gene expression. However, it has also been reported that inhibition of PI3K has no significant effects on Mt expression in immortalized epithelial cells and increases Mt expression in HCC cells. To further characterize the role of the PI3K pathway on the activity of MTF-1, transfection experiments were performed in HEK293 and HepG2 cells in presence of glycogen synthase kinase-3 (GSK-3), mTOR-C1, and mTOR-C2 inhibitors, as well as of siRNAs targeting Phosphatase and TENsin homolog (PTEN). We showed that inhibition of the mTOR-C2 complex inhibits the activity of MTF-1 in HepG2 and HEK293 cells, while inhibition of the mTOR-C1 complex or of PTEN stimulates MTF-1 activity in HEK293 cells. These results confirm that the PI3K pathway positively regulates MTF-1 activity. Finally, we showed that GSK-3 is required for MTF-1 activation in response to zinc ions.

9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL