1
|
Zanela TMP, Zangiabadi M, Zhao Y, Underbakke ES. Molecularly imprinted nanoparticles reveal regulatory scaffolding features in Pyk2 tyrosine kinase. RSC Chem Biol 2024; 5:447-453. [PMID: 38725907 PMCID: PMC11078204 DOI: 10.1039/d3cb00228d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/13/2024] [Indexed: 05/12/2024] Open
Abstract
Pyk2 is a multi-domain non-receptor tyrosine kinase that serves dual roles as a signaling enzyme and scaffold. Pyk2 activation involves a multi-stage cascade of conformational rearrangements and protein interactions initiated by autophosphorylation of a linker site. Linker phosphorylation recruits Src kinase, and Src-mediated phosphorylation of the Pyk2 activation loop confers full activation. The regulation and accessibility of the initial Pyk2 autophosphorylation site remains unclear. We employed peptide-binding molecularly imprinted nanoparticles (MINPs) to probe the regulatory conformations controlling Pyk2 activation. MINPs differentiating local structure and phosphorylation state revealed that the Pyk2 autophosphorylation site is protected in the autoinhibited state. Activity profiling of Pyk2 variants implicated FERM and linker residues responsible for constraining the autophosphorylation site. MINPs targeting each Src docking site disrupt the higher-order kinase interactions critical for activation complex maturation. Ultimately, MINPs targeting key regulatory motifs establish a useful toolkit for probing successive activational stages in the higher-order Pyk2 signaling complex.
Collapse
Affiliation(s)
- Tania M Palhano Zanela
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University Ames IA 50011 USA
| | - Milad Zangiabadi
- Department of Chemistry, Iowa State University Ames Iowa 50011 USA
| | - Yan Zhao
- Department of Chemistry, Iowa State University Ames Iowa 50011 USA
| | - Eric S Underbakke
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University Ames IA 50011 USA
| |
Collapse
|
2
|
López-Molina L, Sancho-Balsells A, Al-Massadi O, Montalban E, Alberch J, Arranz B, Girault JA, Giralt A. Hippocampal Pyk2 regulates specific social skills: Implications for schizophrenia. Neurobiol Dis 2024; 194:106487. [PMID: 38552722 DOI: 10.1016/j.nbd.2024.106487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 04/01/2024] Open
Abstract
Pyk2 has been shown previously to be involved in several psychological and cognitive alterations related to stress, Huntington's disease, and Alzheimer's disease. All these disorders are accompanied by different types of impairments in sociability, which has recently been linked to improper mitochondrial function. We hypothesize that Pyk2, which regulates mitochondria, could be associated with the regulation of mitochondrial dynamics and social skills. In the present manuscript, we report that a reduction of Pyk2 levels in mouse pyramidal neurons of the hippocampus decreased social dominance and aggressivity. Furthermore, social interactions induced robust Pyk2-dependent hippocampal changes in several oxidative phosphorylation complexes. We also observed that Pyk2 levels were increased in the CA1 pyramidal neurons of schizophrenic subjects, occurring alongside changes in different direct and indirect regulators of mitochondrial function including DISC1 and Grp75. Accordingly, overexpressing Pyk2 in hippocampal CA1 pyramidal cells mimicked some specific schizophrenia-like social behaviors in mice. In summary, our results indicate that Pyk2 might play a role in regulating specific social skills likely via mitochondrial dynamics and that there might be a link between Pyk2 levels in hippocampal neurons and social disturbances in schizophrenia.
Collapse
Affiliation(s)
- Laura López-Molina
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Omar Al-Massadi
- Inserm UMR-S 1270, 75005 Paris, France; Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France; Institut du Fer a Moulin, 75005 Paris, France; Translational Endocrinology Group, Servicio de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (IDIS/CHUS), Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Spain
| | - Enrica Montalban
- Inserm UMR-S 1270, 75005 Paris, France; Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France; Institut du Fer a Moulin, 75005 Paris, France; UMR 1286, NutriNeuro - INRAE / Université de Bordeaux / INP 146, rue Léo Saignat, 33076 Brodeaux cedex, France
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Faculty of Medicine and Health Science, Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Belén Arranz
- Parc Sanitari Sant Joan de Déu, CIBERSAM, Barcelona, Spain
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, 75005 Paris, France; Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France; Institut du Fer a Moulin, 75005 Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Faculty of Medicine and Health Science, Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
Lin Y, Yang J, Yang Q, Zeng S, Zhang J, Zhu Y, Tong Y, Li L, Tan W, Chen D, Sun Q. PTK2B promotes TBK1 and STING oligomerization and enhances the STING-TBK1 signaling. Nat Commun 2023; 14:7567. [PMID: 37989995 PMCID: PMC10663505 DOI: 10.1038/s41467-023-43419-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
TANK-binding kinase 1 (TBK1) is a key kinase in regulating antiviral innate immune responses. While the oligomerization of TBK1 is critical for its full activation, the molecular mechanism of how TBK1 forms oligomers remains unclear. Here, we show that protein tyrosine kinase 2 beta (PTK2B) acts as a TBK1-interacting protein and regulates TBK1 oligomerization. Functional assays reveal that PTK2B depletion reduces antiviral signaling in mouse embryonic fibroblasts, macrophages and dendritic cells, and genetic experiments show that Ptk2b-deficient mice are more susceptible to viral infection than control mice. Mechanistically, we demonstrate that PTK2B directly phosphorylates residue Tyr591 of TBK1, which increases TBK1 oligomerization and activation. In addition, we find that PTK2B also interacts with the stimulator of interferon genes (STING) and can promote its oligomerization in a kinase-independent manner. Collectively, PTK2B enhances the oligomerization of TBK1 and STING via different mechanisms, subsequently regulating STING-TBK1 activation to ensure efficient antiviral innate immune responses.
Collapse
Affiliation(s)
- Yongfang Lin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Jia #3 Datun Road, Chaoyang District, 100101, Beijing, China
- Institute of Biomedical Research, Yunnan University, 650500, Kunming, China
- Institute for Stem Cells and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
- School of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Jia #3 Datun Road, Chaoyang District, 100101, Beijing, China
- Institute for Stem Cells and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
- School of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Qili Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Jia #3 Datun Road, Chaoyang District, 100101, Beijing, China
- Institute for Stem Cells and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
- School of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Sha Zeng
- Institute of Biomedical Research, Yunnan University, 650500, Kunming, China
| | - Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Jia #3 Datun Road, Chaoyang District, 100101, Beijing, China
- Institute for Stem Cells and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
- School of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yuanxiang Zhu
- Institute of Biomedical Research, Yunnan University, 650500, Kunming, China
| | - Yuxin Tong
- Institute of Biomedical Research, Yunnan University, 650500, Kunming, China
| | - Lin Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Jia #3 Datun Road, Chaoyang District, 100101, Beijing, China
- Institute for Stem Cells and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Weiqi Tan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Jia #3 Datun Road, Chaoyang District, 100101, Beijing, China
- Institute for Stem Cells and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Dahua Chen
- Institute of Biomedical Research, Yunnan University, 650500, Kunming, China.
| | - Qinmiao Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Jia #3 Datun Road, Chaoyang District, 100101, Beijing, China.
- Institute for Stem Cells and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China.
- School of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
4
|
Hayat A, Carter EP, King HW, Ors A, Doe A, Teijeiro SA, Charrot S, Godinho S, Cutillas P, Mohammed H, Grose RP, Ficz G. Low HER2 expression in normal breast epithelium enables dedifferentiation and malignant transformation via chromatin opening. Dis Model Mech 2023; 16:dmm049894. [PMID: 36661191 PMCID: PMC9922733 DOI: 10.1242/dmm.049894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023] Open
Abstract
Overexpression of the HER2 protein in breast cancer patients is a predictor of poor prognosis and resistance to therapies. We used an inducible breast cancer transformation system that allows investigation of early molecular changes. HER2 overexpression to similar levels as those observed in a subtype of HER2-positive breast cancer patients induced transformation of MCF10A cells and resulted in gross morphological changes, increased anchorage-independent growth of cells, and altered the transcriptional programme of genes associated with oncogenic transformation. Global phosphoproteomic analysis during HER2 induction predominantly detected an increase in protein phosphorylation. Intriguingly, this correlated with chromatin opening, as measured by ATAC-seq on acini isolated from 3D cell culture. HER2 overexpression resulted in opening of many distal regulatory regions and promoted reprogramming-associated heterogeneity. We found that a subset of cells acquired a dedifferentiated breast stem-like phenotype, making them likely candidates for malignant transformation. Our data show that this population of cells, which counterintuitively enriches for relatively low HER2 protein abundance and increased chromatin accessibility, possesses transformational drive, resulting in increased anchorage-independent growth in vitro compared to cells not displaying a stem-like phenotype.
Collapse
Affiliation(s)
- Ateequllah Hayat
- Institute of Medical and Biomedical Education, St George’s, University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Edward P. Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Hamish W. King
- Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical Research, Royal Parade, Parkville, VIC 3052, Australia
| | - Aysegul Ors
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Aaron Doe
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Saul A. Teijeiro
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sarah Charrot
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Susana Godinho
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Pedro Cutillas
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Hisham Mohammed
- Knight Cancer Institute, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Richard P. Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Gabriella Ficz
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
5
|
Oncel S, Basson MD. ZINC40099027 promotes monolayer circular defect closure by a novel pathway involving cytosolic activation of focal adhesion kinase and downstream paxillin and ERK1/2. Cell Tissue Res 2022; 390:261-279. [PMID: 36001146 DOI: 10.1007/s00441-022-03674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/17/2022] [Indexed: 11/02/2022]
Abstract
ZINC40099027 (ZN27) is a specific focal adhesion kinase (FAK) activator that promotes murine mucosal wound closure after ischemic or NSAID-induced injury. Diverse motogenic pathways involve FAK, but the direct consequences of pure FAK activation have not been studied, and how ZN27-induced FAK activation stimulates wound closure remained unclear. We investigated signaling and focal adhesion (FA) turnover after FAK activation by ZN27 in Caco-2 cells, confirming key results in CCD841 cells. ZN27 increased Caco-2 FAK-Y-397, FAK-Y-576/7, paxillin-Y-118, and ERK 1/2 phosphorylation and decreased FAK-Y-925 phosphorylation, without altering FAK-Y-861, p38, Jnk, or Akt phosphorylation. ZN27 increased FAK-paxillin interaction while decreasing FAK-Grb2 association. ZN27 increased membrane-associated FAK-Y-397 and FAK-Y-576/7 phosphorylation and paxillin-Y-118 and ERK 1/2 phosphorylation but decreased FAK-Y-925 phosphorylation without altering Src or Grb2. Moreover, ZN27 increased the fluorescence intensity of GFP-FAK and pFAK-Y397 in FAs and increased the total number of FAs but reduced their size in GFP-FAK-transfected Caco-2 cells, consistent with increased FA turnover. In contrast, FAK-Y397F transfection prevented ZN27 effects on FAK size and number and FAK and pFAK fluorescent intensity in FAs. We confirmed the proposed FAK/paxillin/ERK pathway using PP2 and U0126 to block Src and MEK1/2 in Caco-2 and CCD841 cells. These results suggest that ZN27 promotes intestinal epithelial monolayer defect closure by stimulating autophosphorylation of FAK in the cytosol, distinct from classical models of FAK activation in the FA. Phosphorylated FAK translocates to the membrane, where its downstream substrates paxillin and ERK are phosphorylated, leading to FA turnover and human intestinal epithelial cell migration.
Collapse
Affiliation(s)
- Sema Oncel
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, USA
| | - Marc D Basson
- Department of Biomedical Sciences, Department of Surgery, Department of Pathology, University of North Dakota School of Medicine & Health Sciences, Grand Forks, USA.
| |
Collapse
|
6
|
Momin AA, Mendes T, Barthe P, Faure C, Hong S, Yu P, Kadaré G, Jaremko M, Girault JA, Jaremko Ł, Arold ST. PYK2 senses calcium through a disordered dimerization and calmodulin-binding element. Commun Biol 2022; 5:800. [PMID: 35945264 PMCID: PMC9363500 DOI: 10.1038/s42003-022-03760-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/22/2022] [Indexed: 11/25/2022] Open
Abstract
Multidomain kinases use many ways to integrate and process diverse stimuli. Here, we investigated the mechanism by which the protein tyrosine kinase 2-beta (PYK2) functions as a sensor and effector of cellular calcium influx. We show that the linker between the PYK2 kinase and FAT domains (KFL) encompasses an unusual calmodulin (CaM) binding element. PYK2 KFL is disordered and engages CaM through an ensemble of transient binding events. Calcium increases the association by promoting structural changes in CaM that expose auxiliary interaction opportunities. KFL also forms fuzzy dimers, and dimerization is enhanced by CaM binding. As a monomer, however, KFL associates with the PYK2 FERM-kinase fragment. Thus, we identify a mechanism whereby calcium influx can promote PYK2 self-association, and hence kinase-activating trans-autophosphorylation. Collectively, our findings describe a flexible protein module that expands the paradigms for CaM binding and self-association, and their use for controlling kinase activity. Protein tyrosine kinase 2-beta is shown to function as a sensor and effector of cellular calcium influx through self-association.
Collapse
Affiliation(s)
- Afaque A Momin
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.,Bioscience Program, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Tiago Mendes
- Inserm UMR-S 1270, Sorbonne Université, Faculty of Sciences and Engineering, Institut du Fer à Moulin, 75005, Paris, France
| | - Philippe Barthe
- Centre de Biologie Structurale (CBS), University Montpellier, INSERM U1054, CNRS UMR 5048, 34090, Montpellier, France
| | - Camille Faure
- Inserm UMR-S 1270, Sorbonne Université, Faculty of Sciences and Engineering, Institut du Fer à Moulin, 75005, Paris, France
| | - SeungBeom Hong
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.,Bioscience Program, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Piao Yu
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.,Bioscience Program, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Gress Kadaré
- Inserm UMR-S 1270, Sorbonne Université, Faculty of Sciences and Engineering, Institut du Fer à Moulin, 75005, Paris, France
| | - Mariusz Jaremko
- Bioscience Program, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Sorbonne Université, Faculty of Sciences and Engineering, Institut du Fer à Moulin, 75005, Paris, France
| | - Łukasz Jaremko
- Bioscience Program, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Stefan T Arold
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia. .,Bioscience Program, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia. .,Centre de Biologie Structurale (CBS), University Montpellier, INSERM U1054, CNRS UMR 5048, 34090, Montpellier, France.
| |
Collapse
|
7
|
Michael E, Polydorides S, Archontis G. Computational Design of Peptides with Improved Recognition of the Focal Adhesion Kinase FAT Domain. Methods Mol Biol 2022; 2405:383-402. [PMID: 35298823 DOI: 10.1007/978-1-0716-1855-4_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We describe a two-stage computational protein design (CPD) methodology for the design of peptides binding to the FAT domain of the protein focal adhesion kinase. The first stage involves high-throughput CPD calculations with the Proteus software. The energies of the folded state are described by a physics-based energy function and of the unfolded peptides by a knowledge-based model that reproduces aminoacid compositions consistent with a helicity scale. The obtained sequences are filtered in terms of the affinity and the stability of the complex. In the second stage, design sequences are further evaluated by all-atom molecular dynamics simulations and binding free energy calculations with a molecular mechanics/implicit solvent free energy function.
Collapse
Affiliation(s)
- Eleni Michael
- Department of Physics, University of Cyprus, Nicosia, Cyprus
| | | | | |
Collapse
|
8
|
de Pins B, Mendes T, Giralt A, Girault JA. The Non-receptor Tyrosine Kinase Pyk2 in Brain Function and Neurological and Psychiatric Diseases. Front Synaptic Neurosci 2021; 13:749001. [PMID: 34690733 PMCID: PMC8527176 DOI: 10.3389/fnsyn.2021.749001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pyk2 is a non-receptor tyrosine kinase highly enriched in forebrain neurons. Pyk2 is closely related to focal adhesion kinase (FAK), which plays an important role in sensing cell contacts with extracellular matrix and other extracellular signals controlling adhesion and survival. Pyk2 shares some of FAK’s characteristics including recruitment of Src-family kinases after autophosphorylation, scaffolding by interacting with multiple partners, and activation of downstream signaling pathways. Pyk2, however, has the unique property to respond to increases in intracellular free Ca2+, which triggers its autophosphorylation following stimulation of various receptors including glutamate NMDA receptors. Pyk2 is dephosphorylated by the striatal-enriched phosphatase (STEP) that is highly expressed in the same neuronal populations. Pyk2 localization in neurons is dynamic, and altered following stimulation, with post-synaptic and nuclear enrichment. As a signaling protein Pyk2 is involved in multiple pathways resulting in sometimes opposing functions depending on experimental models. Thus Pyk2 has a dual role on neurites and dendritic spines. With Src family kinases Pyk2 participates in postsynaptic regulations including of NMDA receptors and is necessary for specific types of synaptic plasticity and spatial memory tasks. The diverse functions of Pyk2 are also illustrated by its role in pathology. Pyk2 is activated following epileptic seizures or ischemia-reperfusion and may contribute to the consequences of these insults whereas Pyk2 deficit may contribute to the hippocampal phenotype of Huntington’s disease. Pyk2 gene, PTK2B, is associated with the risk for late-onset Alzheimer’s disease. Studies of underlying mechanisms indicate a complex contribution with involvement in amyloid toxicity and tauopathy, combined with possible functional deficits in neurons and contribution in microglia. A role of Pyk2 has also been proposed in stress-induced depression and cocaine addiction. Pyk2 is also important for the mobility of astrocytes and glioblastoma cells. The implication of Pyk2 in various pathological conditions supports its potential interest for therapeutic interventions. This is possible through molecules inhibiting its activity or increasing it through inhibition of STEP or other means, depending on a precise evaluation of the balance between positive and negative consequences of Pyk2 actions.
Collapse
Affiliation(s)
- Benoit de Pins
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Tiago Mendes
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| |
Collapse
|
9
|
González Wusener AE, González Á, Perez Collado ME, Maza MR, General IJ, Arregui CO. Protein tyrosine phosphatase 1B targets focal adhesion kinase and paxillin in cell-matrix adhesions. J Cell Sci 2021; 134:272564. [PMID: 34553765 DOI: 10.1242/jcs.258769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/14/2021] [Indexed: 11/20/2022] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B, also known as PTPN1) is an established regulator of cell-matrix adhesion and motility. However, the nature of substrate targets at adhesion sites remains to be validated. Here, we used bimolecular fluorescence complementation assays, in combination with a substrate trapping mutant of PTP1B, to directly examine whether relevant phosphotyrosines on paxillin and focal adhesion kinase (FAK, also known as PTK2) are substrates of the phosphatase in the context of cell-matrix adhesion sites. We found that the formation of catalytic complexes at cell-matrix adhesions requires intact tyrosine residues Y31 and Y118 on paxillin, and the localization of FAK at adhesion sites. Additionally, we found that PTP1B specifically targets Y925 on the focal adhesion targeting (FAT) domain of FAK at adhesion sites. Electrostatic analysis indicated that dephosphorylation of this residue promotes the closed conformation of the FAT 4-helix bundle and its interaction with paxillin at adhesion sites.
Collapse
Affiliation(s)
- Ana E González Wusener
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| | - Ángela González
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| | - María E Perez Collado
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| | - Melina R Maza
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martin, Instituto de Ciencias Físicas and CONICET, San Martin, Buenos Aires 1650, Argentina
| | - Ignacio J General
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martin, Instituto de Ciencias Físicas and CONICET, San Martin, Buenos Aires 1650, Argentina
| | - Carlos O Arregui
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| |
Collapse
|
10
|
Pyk2 in dorsal hippocampus plays a selective role in spatial memory and synaptic plasticity. Sci Rep 2021; 11:16357. [PMID: 34381140 PMCID: PMC8358019 DOI: 10.1038/s41598-021-95813-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Pyk2 is a Ca2+-activated non-receptor tyrosine kinase enriched in the forebrain, especially in pyramidal neurons of the hippocampus. Previous reports suggested its role in hippocampal synaptic plasticity and spatial memory but with contradictory findings possibly due to experimental conditions. Here we address this issue and show that novel object location, a simple test of spatial memory induced by a single training session, is altered in Pyk2 KO mice and that re-expression of Pyk2 in the dorsal hippocampus corrects this deficit. Bilateral targeted deletion of Pyk2 in dorsal hippocampus CA1 region also alters novel object location. Long term potentiation (LTP) in CA1 is impaired in Pyk2 KO mice using a high frequency stimulation induction protocol but not with a theta burst protocol, explaining differences between previous reports. The same selective LTP alteration is observed in mice with Pyk2 deletion in dorsal hippocampus CA1 region. Thus, our results establish the role of Pyk2 in specific aspects of spatial memory and synaptic plasticity and show the dependence of the phenotype on the type of experiments used to reveal it. In combination with other studies, we provide evidence for a selective role of non-receptor tyrosine kinases in specific aspects of hippocampal neurons synaptic plasticity.
Collapse
|
11
|
Abstract
FAK, a nonreceptor tyrosine kinase, has been recognized as a novel target class for the development of targeted anticancer agents. Overexpression of FAK is a common occurrence in several solid tumors, in which the kinase has been implicated in promoting metastases. Consequently, designing and developing potent FAK inhibitors is becoming an attractive goal, and FAK inhibitors are being recognized as a promising tool in our armamentarium for treating diverse cancers. This review comprehensively summarizes the different classes of synthetically derived compounds that have been reported as potent FAK inhibitors in the last three decades. Finally, the future of FAK-targeting smart drugs that are designed to slow down the emergence of drug resistance is discussed.
Collapse
|
12
|
Alexander RA, Lot I, Saha K, Abadie G, Lambert M, Decosta E, Kobayashi H, Beautrait A, Borrull A, Asnacios A, Bouvier M, Scott MGH, Marullo S, Enslen H. Beta-arrestins operate an on/off control switch for focal adhesion kinase activity. Cell Mol Life Sci 2020; 77:5259-5279. [PMID: 32040695 PMCID: PMC11104786 DOI: 10.1007/s00018-020-03471-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 12/20/2022]
Abstract
Focal adhesion kinase (FAK) regulates key biological processes downstream of G protein-coupled receptors (GPCRs) in normal and cancer cells, but the modes of kinase activation by these receptors remain unclear. We report that after GPCR stimulation, FAK activation is controlled by a sequence of events depending on the scaffolding proteins β-arrestins and G proteins. Depletion of β-arrestins results in a marked increase in FAK autophosphorylation and focal adhesion number. We demonstrate that β-arrestins interact directly with FAK and inhibit its autophosphorylation in resting cells. Both FAK-β-arrestin interaction and FAK inhibition require the FERM domain of FAK. Following the stimulation of the angiotensin receptor AT1AR and subsequent translocation of the FAK-β-arrestin complex to the plasma membrane, β-arrestin interaction with the adaptor AP-2 releases inactive FAK from the inhibitory complex, allowing its activation by receptor-stimulated G proteins and activation of downstream FAK effectors. Release and activation of FAK in response to angiotensin are prevented by an AP-2-binding deficient β-arrestin and by a specific inhibitor of β-arrestin/AP-2 interaction; this inhibitor also prevents FAK activation in response to vasopressin. This previously unrecognized mechanism of FAK regulation involving a dual role of β-arrestins, which inhibit FAK in resting cells while driving its activation at the plasma membrane by GPCR-stimulated G proteins, opens new potential therapeutic perspectives in cancers with up-regulated FAK.
Collapse
Affiliation(s)
- Revu Ann Alexander
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Isaure Lot
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Kusumika Saha
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Guillaume Abadie
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Mireille Lambert
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Eleonore Decosta
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Hiroyuki Kobayashi
- Department of Biochemistry and the Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Alexandre Beautrait
- Department of Biochemistry and the Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Aurélie Borrull
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Atef Asnacios
- Laboratoire Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, Paris, France
| | - Michel Bouvier
- Department of Biochemistry and the Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Mark G H Scott
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Stefano Marullo
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Hervé Enslen
- Institut Cochin, Inserm U 1016, CNRS UMR8104, Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.
| |
Collapse
|
13
|
Acebrón I, Righetto RD, Schoenherr C, de Buhr S, Redondo P, Culley J, Rodríguez CF, Daday C, Biyani N, Llorca O, Byron A, Chami M, Gräter F, Boskovic J, Frame MC, Stahlberg H, Lietha D. Structural basis of Focal Adhesion Kinase activation on lipid membranes. EMBO J 2020; 39:e104743. [PMID: 32779739 PMCID: PMC7527928 DOI: 10.15252/embj.2020104743] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Focal adhesion kinase (FAK) is a key component of the membrane proximal signaling layer in focal adhesion complexes, regulating important cellular processes, including cell migration, proliferation, and survival. In the cytosol, FAK adopts an autoinhibited state but is activated upon recruitment into focal adhesions, yet how this occurs or what induces structural changes is unknown. Here, we employ cryo-electron microscopy to reveal how FAK associates with lipid membranes and how membrane interactions unlock FAK autoinhibition to promote activation. Intriguingly, initial binding of FAK to the membrane causes steric clashes that release the kinase domain from autoinhibition, allowing it to undergo a large conformational change and interact itself with the membrane in an orientation that places the active site toward the membrane. In this conformation, the autophosphorylation site is exposed and multiple interfaces align to promote FAK oligomerization on the membrane. We show that interfaces responsible for initial dimerization and membrane attachment are essential for FAK autophosphorylation and resulting cellular activity including cancer cell invasion, while stable FAK oligomerization appears to be needed for optimal cancer cell proliferation in an anchorage-independent manner. Together, our data provide structural details of a key membrane bound state of FAK that is primed for efficient autophosphorylation and activation, hence revealing the critical event in integrin mediated FAK activation and signaling at focal adhesions.
Collapse
Affiliation(s)
- Iván Acebrón
- Structural Biology ProgrammeSpanish National Cancer Research CentreMadridSpain
| | - Ricardo D Righetto
- Center for Cellular Imaging and NanoAnalyticsBiozentrumUniversity of BaselBaselSwitzerland
| | - Christina Schoenherr
- Cancer Research UK Edinburgh CentreInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Svenja de Buhr
- Heidelberg Institute for Theoretical StudiesHeidelbergGermany
- Interdisciplinary Center for Scientific ComputingHeidelberg UniversityHeidelbergGermany
| | - Pilar Redondo
- Structural Biology ProgrammeSpanish National Cancer Research CentreMadridSpain
| | - Jayne Culley
- Cancer Research UK Edinburgh CentreInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Carlos F Rodríguez
- Structural Biology ProgrammeSpanish National Cancer Research CentreMadridSpain
| | - Csaba Daday
- Heidelberg Institute for Theoretical StudiesHeidelbergGermany
- Interdisciplinary Center for Scientific ComputingHeidelberg UniversityHeidelbergGermany
| | - Nikhil Biyani
- Center for Cellular Imaging and NanoAnalyticsBiozentrumUniversity of BaselBaselSwitzerland
| | - Oscar Llorca
- Structural Biology ProgrammeSpanish National Cancer Research CentreMadridSpain
| | - Adam Byron
- Cancer Research UK Edinburgh CentreInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Mohamed Chami
- Center for Cellular Imaging and NanoAnalyticsBiozentrumUniversity of BaselBaselSwitzerland
| | - Frauke Gräter
- Heidelberg Institute for Theoretical StudiesHeidelbergGermany
- Interdisciplinary Center for Scientific ComputingHeidelberg UniversityHeidelbergGermany
| | - Jasminka Boskovic
- Structural Biology ProgrammeSpanish National Cancer Research CentreMadridSpain
| | - Margaret C Frame
- Cancer Research UK Edinburgh CentreInstitute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Henning Stahlberg
- Center for Cellular Imaging and NanoAnalyticsBiozentrumUniversity of BaselBaselSwitzerland
| | - Daniel Lietha
- Structural Biology ProgrammeSpanish National Cancer Research CentreMadridSpain
- Centro de Investigaciones Biológicas Margarita SalasSpanish National Research Council (CSIC)MadridSpain
| |
Collapse
|
14
|
Michael E, Polydorides S, Promponas VJ, Skourides P, Archontis G. Recognition of LD motifs by the focal adhesion targeting domains of focal adhesion kinase and proline-rich tyrosine kinase 2-beta: Insights from molecular dynamics simulations. Proteins 2020; 89:29-52. [PMID: 32776636 DOI: 10.1002/prot.25992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/21/2020] [Accepted: 07/26/2020] [Indexed: 12/13/2022]
Abstract
The focal adhesion kinase (FAK) and the proline-rich tyrosine kinase 2-beta (PYK2) are implicated in cancer progression and metastasis and represent promising biomarkers and targets for cancer therapy. FAK and PYK2 are recruited to focal adhesions (FAs) via interactions between their FA targeting (FAT) domains and conserved segments (LD motifs) on the proteins Paxillin, Leupaxin, and Hic-5. A promising new approach for the inhibition of FAK and PYK2 targets interactions of the FAK domains with proteins that promote localization at FAs. Advances toward this goal include the development of surface plasmon resonance, heteronuclear single quantum coherence nuclear magnetic resonance (HSQC-NMR) and fluorescence polarization assays for the identification of fragments or compounds interfering with the FAK-Paxillin interaction. We have recently validated this strategy, showing that Paxillin mimicking polypeptides with 2 to 3 LD motifs displace FAK from FAs and block kinase-dependent and independent functions of FAK, including downstream integrin signaling and FA localization of the protein p130Cas. In the present work we study by all-atom molecular dynamics simulations the recognition of peptides with the Paxillin and Leupaxin LD motifs by the FAK-FAT and PYK2-FAT domains. Our simulations and free-energy analysis interpret experimental data on binding of Paxillin and Leupaxin LD motifs at FAK-FAT and PYK2-FAT binding sites, and assess the roles of consensus LD regions and flanking residues. Our results can assist in the design of effective inhibitory peptides of the FAK-FAT: Paxillin and PYK2-FAT:Leupaxin complexes and the construction of pharmacophore models for the discovery of potential small-molecule inhibitors of the FAK-FAT and PYK2-FAT focal adhesion based functions.
Collapse
Affiliation(s)
- Eleni Michael
- Department of Physics, University of Cyprus, Nicosia, Cyprus
| | | | - Vasilis J Promponas
- Bioinformatics Research Laboratory, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Paris Skourides
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | | |
Collapse
|
15
|
de Pins B, Montalban E, Vanhoutte P, Giralt A, Girault JA. The non-receptor tyrosine kinase Pyk2 modulates acute locomotor effects of cocaine in D1 receptor-expressing neurons of the nucleus accumbens. Sci Rep 2020; 10:6619. [PMID: 32313025 PMCID: PMC7170924 DOI: 10.1038/s41598-020-63426-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/20/2020] [Indexed: 01/16/2023] Open
Abstract
The striatum is critical for cocaine-induced locomotor responses. Although the role of D1 receptor-expressing neurons is established, underlying molecular pathways are not fully understood. We studied the role of Pyk2, a non-receptor, calcium-dependent protein-tyrosine kinase. The locomotor coordination and basal activity of Pyk2 knock-out mice were not altered and major striatal protein markers were normal. Cocaine injection increased Pyk2 tyrosine phosphorylation in mouse striatum. Pyk2-deficient mice displayed decreased locomotor response to acute cocaine injection. In contrast, locomotor sensitization and conditioned place preference were normal. Cocaine-activated ERK phosphorylation, a signaling pathway essential for these late responses, was unaltered. Conditional deletion of Pyk2 in the nucleus accumbens or in D1 neurons reproduced decreased locomotor response to cocaine, whereas deletion of Pyk2 in the dorsal striatum or in A2A receptor-expressing neurons did not. In mice lacking Pyk2 in D1-neurons locomotor response to D1 agonist SKF-81297, but not to an anticholinergic drug, was blunted. Our results identify Pyk2 as a regulator of acute locomotor responses to psychostimulants. They highlight the role of tyrosine phosphorylation pathways in striatal neurons and suggest that changes in Pyk2 expression or activation may alter specific responses to drugs of abuse, or possibly other behavioral responses linked to dopamine action.
Collapse
Affiliation(s)
- Benoit de Pins
- Inserm UMR-S 1270, Paris, 75005, France
- Sorbonne Université, Faculty of Sciences and Engineering, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Enrica Montalban
- Inserm UMR-S 1270, Paris, 75005, France
- Sorbonne Université, Faculty of Sciences and Engineering, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
- BFA - Unité de Biologie Fonctionnelle et Adaptative - CNRS UMR 8251, Paris University, Paris, 75205, France
| | - Peter Vanhoutte
- Sorbonne Université, Faculty of Sciences and Engineering, Paris, 75005, France
- Inserm UMR-S 1130, Neurosciences Paris Seine, Paris, 75005, France
- CNRS UMR 8246, Paris, 75005, France
| | - Albert Giralt
- Inserm UMR-S 1270, Paris, 75005, France
- Sorbonne Université, Faculty of Sciences and Engineering, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, 28031, Spain
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris, 75005, France.
- Sorbonne Université, Faculty of Sciences and Engineering, Paris, 75005, France.
- Institut du Fer à Moulin, Paris, 75005, France.
| |
Collapse
|
16
|
Higa‐Nakamine S, Okitsu‐Sakurayama S, Kina S, Yamamoto H. Fyn‐mediated phosphorylation of Pyk2 promotes its activation and dissociation downstream of gonadotropin‐releasing hormone receptor. FEBS J 2020; 287:3551-3564. [DOI: 10.1111/febs.15231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/13/2019] [Accepted: 01/27/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Sayomi Higa‐Nakamine
- Department of Biochemistry Graduate School of Medicine University of the Ryukyus Okinawa Japan
| | - Shiho Okitsu‐Sakurayama
- Department of Biochemistry Graduate School of Medicine University of the Ryukyus Okinawa Japan
| | - Shinichiro Kina
- Department of Oral and Maxillofacial Functional Rehabilitation Graduate School of Medicine University of the Ryukyus Okinawa Japan
| | - Hideyuki Yamamoto
- Department of Biochemistry Graduate School of Medicine University of the Ryukyus Okinawa Japan
| |
Collapse
|
17
|
Afonso P, De Luca P, Carvalho RS, Cortes L, Pinheiro P, Oliveiros B, Almeida RD, Mele M, Duarte CB. BDNF increases synaptic NMDA receptor abundance by enhancing the local translation of Pyk2 in cultured hippocampal neurons. Sci Signal 2019; 12:12/586/eaav3577. [PMID: 31213568 DOI: 10.1126/scisignal.aav3577] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The effects of brain-derived neurotrophic factor (BDNF) in long-term synaptic potentiation (LTP) are thought to underlie learning and memory formation and are partly mediated by local protein synthesis. Here, we investigated the mechanisms that mediate BDNF-induced alterations in the synaptic proteome that are coupled to synaptic strengthening. BDNF induced the synaptic accumulation of GluN2B-containing NMDA receptors (NMDARs) and increased the amplitude of NMDAR-mediated miniature excitatory postsynaptic currents (mEPSCs) in cultured rat hippocampal neurons by a mechanism requiring activation of the protein tyrosine kinase Pyk2 and dependent on cellular protein synthesis. Single-particle tracking using quantum dot imaging revealed that the increase in the abundance of synaptic NMDAR currents correlated with their enhanced stability in the synaptic compartment. Furthermore, BDNF increased the local synthesis of Pyk2 at the synapse, and the observed increase in Pyk2 protein abundance along dendrites of cultured hippocampal neurons was mediated by a mechanism dependent on the ribonucleoprotein hnRNP K, which bound to Pyk2 mRNA and dissociated from it upon BDNF application. Knocking down hnRNP K reduced the BDNF-induced synaptic synthesis of Pyk2 protein, whereas its overexpression enhanced it. Together, these findings indicate that hnRNP K mediates the synaptic distribution of Pyk2 synthesis, and hence the synaptic incorporation of GluN2B-containing NMDARs, induced by BDNF, which may affect LTP and synaptic plasticity.
Collapse
Affiliation(s)
- Pedro Afonso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Pasqualino De Luca
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-790 Coimbra, Portugal
| | - Rafael S Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Luísa Cortes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-790 Coimbra, Portugal
| | - Paulo Pinheiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-790 Coimbra, Portugal
| | - Barbara Oliveiros
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ramiro D Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Health Sciences Program, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Miranda Mele
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-790 Coimbra, Portugal
| | - Carlos B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal. .,Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| |
Collapse
|
18
|
Naser R, Aldehaiman A, Díaz-Galicia E, Arold ST. Endogenous Control Mechanisms of FAK and PYK2 and Their Relevance to Cancer Development. Cancers (Basel) 2018; 10:E196. [PMID: 29891810 PMCID: PMC6025627 DOI: 10.3390/cancers10060196] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/31/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023] Open
Abstract
Focal adhesion kinase (FAK) and its close paralogue, proline-rich tyrosine kinase 2 (PYK2), are key regulators of aggressive spreading and metastasis of cancer cells. While targeted small-molecule inhibitors of FAK and PYK2 have been found to have promising antitumor activity, their clinical long-term efficacy may be undermined by the strong capacity of cancer cells to evade anti-kinase drugs. In healthy cells, the expression and/or function of FAK and PYK2 is tightly controlled via modulation of gene expression, competing alternatively spliced forms, non-coding RNAs, and proteins that directly or indirectly affect kinase activation or protein stability. The molecular factors involved in this control are frequently deregulated in cancer cells. Here, we review the endogenous mechanisms controlling FAK and PYK2, and with particular focus on how these mechanisms could inspire or improve anticancer therapies.
Collapse
Affiliation(s)
- Rayan Naser
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Abdullah Aldehaiman
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Escarlet Díaz-Galicia
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| |
Collapse
|
19
|
PTK2B/Pyk2 overexpression improves a mouse model of Alzheimer's disease. Exp Neurol 2018; 307:62-73. [PMID: 29803828 DOI: 10.1016/j.expneurol.2018.05.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/14/2018] [Accepted: 05/23/2018] [Indexed: 12/31/2022]
Abstract
Pyk2 is a Ca2+-activated non-receptor tyrosine kinase enriched in forebrain neurons and involved in synaptic regulation. Human genetic studies associated PTK2B, the gene coding Pyk2, with risk for Alzheimer's disease (AD). We previously showed that Pyk2 is important for hippocampal function, plasticity, and spine structure. However, its potential role in AD is unknown. To address this question we used human brain samples and 5XFAD mice, an amyloid mouse model of AD expressing mutated human amyloid precursor protein and presenilin1. In the hippocampus of 5XFAD mice and in human AD patients' cortex and hippocampus, Pyk2 total levels were normal. However, Pyk2 Tyr-402 phosphorylation levels, reflecting its autophosphorylation-dependent activity, were reduced in 5XFAD mice at 8 months of age but not 3 months. We crossed these mice with Pyk2-/- mice to generate 5XFAD animals devoid of Pyk2. At 8 months the phenotype of 5XFAD x Pyk2-/- double mutant mice was not different from that of 5XFAD. In contrast, overexpression of Pyk2 in the hippocampus of 5XFAD mice, using adeno-associated virus, rescued autophosphorylated Pyk2 levels and improved synaptic markers and performance in several behavioral tasks. Both Pyk2-/- and 5XFAD mice showed an increase of potentially neurotoxic Src cleavage product, which was rescued by Pyk2 overexpression. Manipulating Pyk2 levels had only minor effects on Aβ plaques, which were slightly decreased in hippocampus CA3 region of double mutant mice and increased following overexpression. Our results show that Pyk2 is not essential for the pathogenic effects of human amyloidogenic mutations in the 5XFAD mouse model. However, the slight decrease in plaque number observed in these mice in the absence of Pyk2 and their increase following Pyk2 overexpression suggest a contribution of this kinase in plaque formation. Importantly, a decreased function of Pyk2 was observed in 5XFAD mice, indicated by its decreased autophosphorylation and associated Src alterations. Overcoming this deficit by Pyk2 overexpression improved the behavioral and molecular phenotype of 5XFAD mice. Thus, our results in a mouse model of AD suggest that Pyk2 impairment may play a role in the symptoms of the disease.
Collapse
|
20
|
Mohanty P, Bhatnagar S. Structure of focal adhesion kinase in healthy heart versus pathological cardiac hypertrophy: A modeling and simulation study. J Mol Graph Model 2017; 80:15-24. [PMID: 29306139 DOI: 10.1016/j.jmgm.2017.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022]
Abstract
Focal adhesion kinase (FAK) is required for signaling in the heart. S910 phosphorylated FAK is known to cause pathological cardiac hypertrophy. The switching of FAK between its inactive (-i), activated (-a) and hyperactive (-h) state is controlled by phosphorylation. FAK consists of three domains, namely: FERM, Kinase, and FAT joined by linkers L1 and L2. The structural basis of FAK phosphorylation and signaling to the downstream pathways is not understood. In this work, we carried out homology modeling and domain assembly of full length human iFAK and aFAK. 100 ns classical molecular dynamic simulations were performed using AMBER14 and effect of S910 phosphorylation on FAK was investigated. The iFAK model superposed on a small angel X-ray scattering (SAXS) derived model with RMSD of 1.18 Å for 590 Cα atoms. aFAK showed S910 phosphorylation site in L2 shielded by FERM. S910 phosphorylation in hFAK led to its exposure accompanied by a large conformational change and exposing the previously buried Grb2 interaction site responsible for causing cardiac hypertrophy. The models of FAK are in agreement with diverse experimental data and observed differences in biological action. Understanding the structure activity relationships of FAK in response to phosphorylation is important for its future therapeutic modulation.
Collapse
Affiliation(s)
- Pallavi Mohanty
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi 110078, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi 110078, India.
| |
Collapse
|
21
|
Zak TJ, Koshman YE, Samarel AM, Robia SL. Regulation of Focal Adhesion Kinase through a Direct Interaction with an Endogenous Inhibitor. Biochemistry 2017; 56:4722-4731. [PMID: 28782937 DOI: 10.1021/acs.biochem.7b00616] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Focal adhesion kinase (FAK) plays a key role in integrin and growth factor signaling pathways. FAK-related non-kinase (FRNK) is an endogenous inhibitor of FAK that shares its primary structure with the C-terminal third of FAK. FAK S910 phosphorylation is known to regulate FAK protein-protein interactions, but the role of the equivalent site on FRNK (S217) is unknown. Here we determined that S217 is highly phosphorylated by ERK in cultured rat aortic smooth muscle cells. Blocking phosphorylation by mutation (S217A) greatly increased FRNK inhibitory potency, resulting in strong inhibition of FAK autophosphorylation at Y397 and induction of smooth muscle cell apoptosis. FRNK has been proposed to compete for FAK anchoring sites in focal adhesions, but we did not detect displacement of FAK by WT-FRNK or superinhibitory S217A-FRNK. Instead, we found FRNK interacted directly with FAK, and this interaction is markedly strengthened for the superinhibitory S217A-FRNK. The results suggest that FRNK limits growth and survival signaling pathways by binding directly to FAK in an inhibitory complex, and this inhibition is relieved by phosphorylation of FRNK at S217.
Collapse
Affiliation(s)
- Taylor J Zak
- Department of Cell and Molecular Physiology and ‡Department of Medicine, Cardiovascular Research Institute, Stritch School of Medicine, Loyola University Chicago , Maywood, Illinois 60153, United States
| | - Yevgenia E Koshman
- Department of Cell and Molecular Physiology and ‡Department of Medicine, Cardiovascular Research Institute, Stritch School of Medicine, Loyola University Chicago , Maywood, Illinois 60153, United States
| | - Allen M Samarel
- Department of Cell and Molecular Physiology and ‡Department of Medicine, Cardiovascular Research Institute, Stritch School of Medicine, Loyola University Chicago , Maywood, Illinois 60153, United States
| | - Seth L Robia
- Department of Cell and Molecular Physiology and ‡Department of Medicine, Cardiovascular Research Institute, Stritch School of Medicine, Loyola University Chicago , Maywood, Illinois 60153, United States
| |
Collapse
|
22
|
Wang H, Luo J, Carlton C, McGinnis LK, Kinsey WH. Sperm-oocyte contact induces outside-in signaling via PYK2 activation. Dev Biol 2017; 428:52-62. [PMID: 28527703 PMCID: PMC5539980 DOI: 10.1016/j.ydbio.2017.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 01/20/2023]
Abstract
Fertilization is a multi-step process that begins with plasma membrane interactions that enable sperm - oocyte binding followed by fusion of the sperm and oocyte plasma membranes. Once membrane fusion has occurred, sperm incorporation involves actin remodeling events within the oocyte cortex that allow the sperm head to penetrate the cortical actin layer and gain access to the ooplasm. Despite the significance for reproduction, the control mechanisms involved in gamete binding, fusion, and sperm incorporation are poorly understood. While it is known that proline - rich tyrosine kinase 2 (PYK2 or PTK2b) kinase activity plays an important role in fertilization, its specific function has not been addressed. The present study made use of a zona-free mouse oocyte fertilization assay to investigate the relationship between PYK2 activity and sperm - oocyte binding and fusion, as well as localized changes in actin polymerization and sperm incorporation. In this assay, the majority of bound sperm had no apparent effect on the oocyte and only a few became incorporated into the ooplasm. However, a subset of bound sperm were associated with a localized response in which PYK2 was recruited to the oocyte cortex where it frequently co-localized with a ring or disk of f-actin. The frequency of sperm-oocyte binding sites that exhibited this actin response was reduced in pyk2-/- oocytes and the pyk2-/- oocytes proved less efficient at incorporating sperm, indicating that this protein kinase may have an important role in sperm incorporation. The response of PYK2 to sperm-oocyte interaction appeared unrelated to gamete fusion since PYK2 was recruited to sperm - binding sites under conditions where sperm - oocyte fusion was prevented and since PYK2 suppression or ablation did not prevent sperm - oocyte fusion. While a direct correlation between the PYK2 response in the oocyte and the successful incorporation of individual bound sperm remains to be established, these findings suggest a model in which the oocyte is not a passive participant in fertilization, but instead responds to sperm contact by localized PYK2 signaling that promotes actin remodeling events required to physically incorporate the sperm head into the ooplasm.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jinping Luo
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA; Applied StemCell Inc., Milpitas, CA 95035, USA
| | - Carol Carlton
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Lynda K McGinnis
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA 90033, USA
| | - William H Kinsey
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
23
|
Pyk2 modulates hippocampal excitatory synapses and contributes to cognitive deficits in a Huntington's disease model. Nat Commun 2017; 8:15592. [PMID: 28555636 PMCID: PMC5459995 DOI: 10.1038/ncomms15592] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
The structure and function of spines and excitatory synapses are under the dynamic control of multiple signalling networks. Although tyrosine phosphorylation is involved, its regulation and importance are not well understood. Here we study the role of Pyk2, a non-receptor calcium-dependent protein-tyrosine kinase highly expressed in the hippocampus. Hippocampal-related learning and CA1 long-term potentiation are severely impaired in Pyk2-deficient mice and are associated with alterations in NMDA receptors, PSD-95 and dendritic spines. In cultured hippocampal neurons, Pyk2 has autophosphorylation-dependent and -independent roles in determining PSD-95 enrichment and spines density. Pyk2 levels are decreased in the hippocampus of individuals with Huntington and in the R6/1 mouse model of the disease. Normalizing Pyk2 levels in the hippocampus of R6/1 mice rescues memory deficits, spines pathology and PSD-95 localization. Our results reveal a role for Pyk2 in spine structure and synaptic function, and suggest that its deficit contributes to Huntington's disease cognitive impairments.
Collapse
|
24
|
Grossi M, Bhattachariya A, Nordström I, Turczyńska KM, Svensson D, Albinsson S, Nilsson BO, Hellstrand P. Pyk2 inhibition promotes contractile differentiation in arterial smooth muscle. J Cell Physiol 2017; 232:3088-3102. [PMID: 28019664 DOI: 10.1002/jcp.25760] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 01/12/2023]
Abstract
Modulation from contractile to synthetic phenotype of vascular smooth muscle cells is a central process in disorders involving compromised integrity of the vascular wall. Phenotype modulation has been shown to include transition from voltage-dependent toward voltage-independent regulation of the intracellular calcium level, and inhibition of non-voltage dependent calcium influx contributes to maintenance of the contractile phenotype. One possible mediator of calcium-dependent signaling is the FAK-family non-receptor protein kinase Pyk2, which is activated by a number of stimuli in a calcium-dependent manner. We used the Pyk2 inhibitor PF-4594755 and Pyk2 siRNA to investigate the role of Pyk2 in phenotype modulation in rat carotid artery smooth muscle cells and in cultured intact arteries. Pyk2 inhibition promoted the expression of smooth muscle markers at the mRNA and protein levels under stimulation by FBS or PDGF-BB and counteracted phenotype shift in cultured intact carotid arteries and balloon injury ex vivo. During long-term (24-96 hr) treatment with PF-4594755, smooth muscle markers increased before cell proliferation was inhibited, correlating with decreased KLF4 expression and differing from effects of MEK inhibition. The Pyk2 inhibitor reduced Orai1 and preserved SERCA2a expression in carotid artery segments in organ culture, and eliminated the inhibitory effect of PDGF stimulation on L-type calcium channel and large-conductance calcium-activated potassium channel expression in carotid cells. Basal intracellular calcium level, calcium wave activity, and store-operated calcium influx were reduced after Pyk2 inhibition of growth-stimulated cells. Pyk2 inhibition may provide an interesting approach for preserving vascular smooth muscle differentiation under pathophysiological conditions.
Collapse
Affiliation(s)
- Mario Grossi
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Ina Nordström
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Daniel Svensson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Bengt-Olof Nilsson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Hall JE, Schaller MD. Phospholipid binding to the FAK catalytic domain impacts function. PLoS One 2017; 12:e0172136. [PMID: 28222177 PMCID: PMC5319746 DOI: 10.1371/journal.pone.0172136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/31/2017] [Indexed: 12/26/2022] Open
Abstract
Focal adhesion kinase is an essential nonreceptor tyrosine kinase that plays an important role in development, in homeostasis and in the progression of human disease. Multiple stimuli activate FAK, which requires a change in structure from an autoinhibited to activated conformation. In the autoinhibited conformation the FERM domain associates with the catalytic domain of FAK and PI(4,5)P2 binding to the FERM domain plays a role in the release of autoinhibition, activating the enzyme. An in silico model of FAK/PI(4,5)P2 interaction suggests that residues on the catalytic domain interact with PI(4,5)P2, in addition to the known FERM domain PI(4,5)P2 binding site. This study was undertaken to test the significance of this in silico observation. Mutations designed to disrupt the putative PI(4,5)P2 binding site were engineered into FAK. These mutants exhibited defects in phosphorylation and failed to completely rescue the phenotype associated with fak-/- phenotype fibroblasts demonstrating the importance of these residues in FAK function. The catalytic domain of FAK exhibited PI(4,5)P2 binding in vitro and binding activity was lost upon mutation of putative PI(4,5)P2 binding site basic residues. However, binding was not selective for PI(4,5)P2, and the catalytic domain bound to several phosphatidylinositol phosphorylation variants. The mutant exhibiting the most severe biological defect was defective for phosphatidylinositol phosphate binding, supporting the model that catalytic domain phospholipid binding is important for biochemical and biological function.
Collapse
Affiliation(s)
- Jessica E. Hall
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia, United States of America
| | - Michael D. Schaller
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia, United States of America
- Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, West Virginia, United States of America
- * E-mail:
| |
Collapse
|
26
|
Kleinschmidt EG, Schlaepfer DD. Focal adhesion kinase signaling in unexpected places. Curr Opin Cell Biol 2017; 45:24-30. [PMID: 28213315 DOI: 10.1016/j.ceb.2017.01.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023]
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase first identified at extracellular matrix and integrin receptor cell adhesion sites and is a key regulator of cell movement. FAK is activated by a variety of stimuli. Herein, we discuss advances in conformational-associated FAK activation and dimerization mechanisms. Additionally, new roles have emerged for FAK signaling at cell adhesions, adherens junctions, endosomes, and the nucleus. In light of these new findings, we review how FAK activation at these sites is connected to the regulation of integrin recycling-activation, vascular permeability, cell survival, and transcriptional regulation, respectively. Studies uncovering FAK signaling connections in unexpected places within cells have yielded important new regulatory insights in cell biology.
Collapse
Affiliation(s)
- Elizabeth G Kleinschmidt
- Biomedical Sciences Graduate Program, University of California, San Diego, CA, United States; Moores Cancer Center, Department of Reproductive Medicine, 3855 Health Sciences Drive, MC 0983, La Jolla, CA 92093-0983, United States
| | - David D Schlaepfer
- Biomedical Sciences Graduate Program, University of California, San Diego, CA, United States; Moores Cancer Center, Department of Reproductive Medicine, 3855 Health Sciences Drive, MC 0983, La Jolla, CA 92093-0983, United States.
| |
Collapse
|
27
|
Endosomes: Emerging Platforms for Integrin-Mediated FAK Signalling. Trends Cell Biol 2016; 26:391-398. [PMID: 26944773 DOI: 10.1016/j.tcb.2016.02.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/22/2016] [Accepted: 02/01/2016] [Indexed: 11/20/2022]
Abstract
Integrins are vital cell adhesion receptors with the ability to transmit extracellular matrix (ECM) cues to intracellular signalling pathways. ECM-integrin signalling regulates various cellular functions such as cell survival and movement. Integrin signalling has been considered to occur exclusively from adhesion sites at the plasma membrane (PM). However, recent data demonstrates integrin signalling also from endosomes. Integrin-mediated focal adhesion kinase (FAK) signalling is strongly dependent on integrin endocytosis, and endosomal FAK signalling facilitates cancer metastasis by supporting anchorage-independent growth and anoikis resistance. Here we discuss the possible mechanisms and functions of endosomal FAK signalling compared with its previously known roles in other cellular locations and discuss the potential of endosomal FAK as novel target for future cancer therapies.
Collapse
|