Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 299(5): 104582, 2023 05.
Article in English | MEDLINE | ID: mdl-36871762

ABSTRACT

The ability to define functional interactions between enzymes and their substrates is crucial for understanding biological control mechanisms; however, such methods face challenges in the transient nature and low stoichiometry of enzyme-substrate interactions. Now, we have developed an optimized strategy that couples substrate-trapping mutagenesis to proximity-labeling mass spectrometry for quantitative analysis of protein complexes involving the protein tyrosine phosphatase PTP1B. This methodology represents a significant shift from classical schemes; it is capable of being performed at near-endogenous expression levels and increasing stoichiometry of target enrichment without a requirement for stimulation of supraphysiological tyrosine phosphorylation levels or maintenance of substrate complexes during lysis and enrichment procedures. Advantages of this new approach are illustrated through application to PTP1B interaction networks in models of HER2-positive and Herceptin-resistant breast cancer. We have demonstrated that inhibitors of PTP1B significantly reduced proliferation and viability in cell-based models of acquired and de novo Herceptin resistance in HER2-positive breast cancer. Using differential analysis, comparing substrate-trapping to wild-type PTP1B, we have identified multiple unreported protein targets of PTP1B with established links to HER2-induced signaling and provided internal validation of method specificity through overlap with previously identified substrate candidates. Overall, this versatile approach can be readily integrated with evolving proximity-labeling platforms (TurboID, BioID2, etc.), and is broadly applicable across all PTP family members for the identification of conditional substrate specificities and signaling nodes in models of human disease.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Signal Transduction , Female , Humans , Breast Neoplasms/genetics , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Trastuzumab/pharmacology , Protein Interaction Mapping
2.
Neurooncol Adv ; 4(1): vdac072, 2022.
Article in English | MEDLINE | ID: mdl-35855490

ABSTRACT

Background: Neurofibromatosis type 2 (NF2) is an autosomal dominant genetic disease characterized by development of schwannomas on the VIIIth (vestibular) cranial nerves. Bromodomain and extra-terminal domain (BET) proteins regulate gene transcription and their activity is required in a variety of cancers including malignant peripheral nerve sheath tumors. The use of BET inhibitors as a therapeutic option to treat NF2 schwannomas has not been explored and is the focus of this study. Methods: A panel of normal and NF2-null Schwann and schwannoma cell lines were used to characterize the impact of the BET inhibitor JQ1 in vitro and in vivo. The mechanism of action was explored by chromatin immunoprecipitation of the BET BRD4, phospho-kinase arrays and immunohistochemistry (IHC) of BRD4 in vestibular schwannomas. Results: JQ1 inhibited proliferation of NF2-null schwannoma and Schwann cell lines in vitro and in vivo. Further, loss of NF2 by CRISPR deletion or siRNA knockdown increased sensitivity of cells to JQ1. Loss of function experiments identified BRD4, and to a lesser extent BRD2, as BET family members mediating the majority of JQ1 effects. IHC demonstrated elevated levels of BRD4 protein in human vestibular schwannomas. Analysis of signaling pathways effected by JQ1 treatment suggests that the effects of JQ1 treatment are mediated, at least in part, via inhibition of PI3K/Akt signaling. Conclusions: NF2-deficient Schwann and schwannoma cells are sensitive to BET inhibition, primarily mediated by BRD4, which is overexpressed in human vestibular schwannomas. Our results suggest BRD4 regulates PI3K signaling and likely impedes NF2 schwannoma growth via this inhibition. These findings implicate BET inhibition as a therapeutic option for NF2-deficient schwannomas.

3.
J Biol Chem ; 294(35): 12992-13005, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31296571

ABSTRACT

Although Merlin's function as a tumor suppressor and regulator of mitogenic signaling networks such as the Ras/rac, Akt, and Hippo pathways is well-documented, in mammals as well as in insects, its role during cell cycle progression remains unclear. In this study, using a combination of approaches, including FACS analysis, time-lapse imaging, immunofluorescence microscopy, and co-immunoprecipitation, we show that Ser-518 of Merlin is a substrate of the Aurora protein kinase A during mitosis and that its phosphorylation facilitates the phosphorylation of a newly discovered site, Thr-581. We found that the expression in HeLa cells of a Merlin variant that is phosphorylation-defective on both sites leads to a defect in centrosomes and mitotic spindles positioning during metaphase and delays the transition from metaphase to anaphase. We also show that the dual mitotic phosphorylation not only reduces Merlin binding to microtubules but also timely modulates ezrin interaction with the cytoskeleton. Finally, we identify several point mutants of Merlin associated with neurofibromatosis type 2 that display an aberrant phosphorylation profile along with defective α-tubulin-binding properties. Altogether, our findings of an Aurora A-mediated interaction of Merlin with α-tubulin and ezrin suggest a potential role for Merlin in cell cycle progression.


Subject(s)
Aurora Kinase A/metabolism , Mitosis , Neurofibromin 2/metabolism , Aurora Kinase A/antagonists & inhibitors , Benzazepines/pharmacology , HEK293 Cells , HeLa Cells , Humans , Mitosis/drug effects , Mutation , Neurofibromin 2/antagonists & inhibitors , Neurofibromin 2/genetics , Nocodazole/pharmacology , Phosphorylation/drug effects
4.
Nat Commun ; 9(1): 1338, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29626191

ABSTRACT

Neurofibromatosis type 2 (NF2) is a tumor-forming disease of the nervous system caused by deletion or by loss-of-function mutations in NF2, encoding the tumor suppressing protein neurofibromin 2 (also known as schwannomin or merlin). Neurofibromin 2 is a member of the ezrin, radixin, moesin (ERM) family of proteins regulating the cytoskeleton and cell signaling. The correlation of the tumor-suppressive function and conformation (open or closed) of neurofibromin 2 has been subject to much speculation, often based on extrapolation from other ERM proteins, and controversy. Here we show that lipid binding results in the open conformation of neurofibromin 2 and that lipid binding is necessary for inhibiting cell proliferation. Collectively, our results provide a mechanism in which the open conformation is unambiguously correlated with lipid binding and localization to the membrane, which are critical for the tumor-suppressive function of neurofibromin 2, thus finally reconciling the long-standing conformation and function debate.


Subject(s)
Lipid Metabolism , Neurofibromin 2/chemistry , Neurofibromin 2/metabolism , Cell Line , Cell Proliferation , Crystallography, X-Ray , HEK293 Cells , Humans , Neurofibromin 2/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Signal Transduction
5.
J Biol Chem ; 292(47): 19179-19197, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28972170

ABSTRACT

Cell adhesion to the extracellular matrix or to surrounding cells plays a key role in cell proliferation and differentiation and is critical for proper tissue homeostasis. An important pathway in adhesion-dependent cell proliferation is the Hippo signaling cascade, which is coregulated by the transcription factors Yes-associated protein 1 (YAP1) and transcriptional coactivator with PDZ-binding motif (TAZ). However, how cells integrate extracellular information at the molecular level to regulate YAP1's nuclear localization is still puzzling. Herein, we investigated the role of ß1 integrins in regulating this process. We found that ß1 integrin-dependent cell adhesion is critical for supporting cell proliferation in mesenchymal cells both in vivo and in vitro ß1 integrin-dependent cell adhesion relied on the relocation of YAP1 to the nucleus after the down-regulation of its phosphorylated state mediated by large tumor suppressor gene 1 and 2 (LATS1/2). We also found that this phenotype relies on ß1 integrin-dependent local activation of the small GTPase RAC1 at the plasma membrane to control the activity of P21 (RAC1)-activated kinase (PAK) of group 1. We further report that the regulatory protein merlin (neurofibromin 2, NF2) interacts with both YAP1 and LATS1/2 via its C-terminal moiety and FERM domain, respectively. PAK1-mediated merlin phosphorylation on Ser-518 reduced merlin's interactions with both LATS1/2 and YAP1, resulting in YAP1 dephosphorylation and nuclear shuttling. Our results highlight RAC/PAK1 as major players in YAP1 regulation triggered by cell adhesion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Genes, Neurofibromatosis 2/physiology , Integrin beta1/physiology , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Neurofibromin 2/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , p21-Activated Kinases/genetics , rac1 GTP-Binding Protein/genetics
6.
Elife ; 62017 05 03.
Article in English | MEDLINE | ID: mdl-28464980

ABSTRACT

The Hippo-YAP pathway is a central regulator of cell contact inhibition, proliferation and death. There are conflicting reports regarding the role of Angiomotin (Amot) in regulating this pathway. While some studies suggest a YAP-inhibitory function other studies indicate Amot is required for YAP activity. Here, we describe an Amot-dependent complex comprised of Amot, YAP and Merlin. The phosphorylation of Amot at Serine 176 shifts localization of this complex to the plasma membrane, where it associates with the tight-junction proteins Pals1/PATJ and E-cadherin. Conversely, hypophosphorylated Amot shifts localization of the complex to the nucleus, where it facilitates the association of YAP and TEAD, induces transcriptional activation of YAP target genes and promotes YAP-dependent cell proliferation. We propose that phosphorylation of AmotS176 is a critical post-translational modification that suppresses YAP's ability to promote cell proliferation and tumorigenesis by altering the subcellular localization of an essential YAP co-factor.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Angiomotins , Cell Membrane/chemistry , Cell Nucleus/chemistry , HEK293 Cells , Hep G2 Cells , Humans , Microfilament Proteins , Phosphorylation , Protein Binding , Transcription Factors , YAP-Signaling Proteins
7.
Cancer Res ; 76(12): 3507-19, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27216189

ABSTRACT

The Hippo-YAP pathway has emerged as a major driver of tumorigenesis in many human cancers. YAP is a transcriptional coactivator and while details of YAP regulation are quickly emerging, it remains unknown what downstream targets are critical for the oncogenic functions of YAP. To determine the mechanisms involved and to identify disease-relevant targets, we examined the role of YAP in neurofibromatosis type 2 (NF2) using cell and animal models. We found that YAP function is required for NF2-null Schwann cell survival, proliferation, and tumor growth in vivo Moreover, YAP promotes transcription of several targets including PTGS2, which codes for COX-2, a key enzyme in prostaglandin biosynthesis, and AREG, which codes for the EGFR ligand, amphiregulin. Both AREG and prostaglandin E2 converge to activate signaling through EGFR. Importantly, treatment with the COX-2 inhibitor celecoxib significantly inhibited the growth of NF2-null Schwann cells and tumor growth in a mouse model of NF2. Cancer Res; 76(12); 3507-19. ©2016 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cyclooxygenase 2/physiology , ErbB Receptors/physiology , Neurofibromatosis 2/etiology , Phosphoproteins/physiology , Signal Transduction/physiology , Amphiregulin/physiology , Carcinogenesis , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Schwann Cells/physiology , Transcription Factors , YAP-Signaling Proteins
8.
Neoplasia ; 18(1): 10-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26806348

ABSTRACT

The Hippo signaling network is a key regulator of cell fate. In the recent years, it was shown that its implication in cancer goes well beyond the sole role of YAP transcriptional activity and its regulation by the canonical MST/LATS kinase cascade. Here we show that the motin family member AMOTL1 is an important effector of Hippo signaling in breast cancer. AMOTL1 connects Hippo signaling to tumor cell aggressiveness. We show that both canonical and noncanonical Hippo signaling modulates AMOTL1 levels. The tumor suppressor Merlin triggers AMOTL1 proteasomal degradation mediated by the NEDD family of ubiquitin ligases through direct interaction. In parallel, YAP stimulates AMOTL1 expression. The loss of Merlin expression and the induction of Yap activity that are frequently observed in breast cancers thus result in elevated AMOTL1 levels. AMOTL1 expression is sufficient to trigger tumor cell migration and stimulates proliferation by activating c-Src. In a large cohort of human breast tumors, we show that AMOTL1 protein levels are upregulated during cancer progression and that, importantly, the expression of AMOTL1 in lymph node metastasis appears predictive of the risk of relapse. Hence we uncover an important mechanism by which Hippo signaling promotes breast cancer progression by modulating the expression of AMOTL1.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Membrane Proteins/metabolism , Neurofibromin 2/metabolism , Angiomotins , Animals , Breast Neoplasms/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Neurofibromin 2/genetics , Nuclear Proteins/metabolism , Protein Binding , Proteolysis , Signal Transduction , Transcription Factors/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...