105 results on '"Edward Manser"'
Search Results
2. Proximity proteomics identifies PAK4 as a component of Afadin–Nectin junctions
- Author
-
Yohendran Baskaran, Felicia Pei-Ling Tay, Elsa Yuen Wai Ng, Claire Lee Foon Swa, Sheena Wee, Jayantha Gunaratne, and Edward Manser
- Subjects
Science - Abstract
PAK4 is a kinase involved in cell-cell junctions, though the identify of the local protein network involving PAK4 is unclear. Here, the authors performed proximity proteomic analysis on mammalian PAK4 and find that PAK4 is associated with Afadin-dependent junctions, and report putative PAK4 phosphorylation substrates at this site.
- Published
- 2021
- Full Text
- View/download PDF
3. Rho GTPases and regulation of cell migration and polarization in human corneal epithelial cells.
- Author
-
Aihua Hou, Li Xian Toh, Kah Hui Gan, Khee Jin Ryan Lee, Edward Manser, and Louis Tong
- Subjects
Medicine ,Science - Abstract
PURPOSE:Epithelial cell migration is required for regeneration of tissues and can be defective in a number of ocular surface diseases. This study aimed to determine the expression pattern of Rho family small G-proteins in human corneal epithelial cells to test their requirement in directional cell migration. METHODS:Rho family small G-protein expression was assessed by reverse transcription-polymerase chain reaction. Dominant-inhibitory constructs encoding Rho proteins or Rho protein targeting small interfering RNA were transfected into human corneal epithelial large T antigen cells, and wound closure rate were evaluated by scratch wounding assay, and a complementary non-traumatic cell migration assay. Immunofluorescence staining was performed to study cell polarization and to assess Cdc42 downstream effector. RESULTS:Cdc42, Chp, Rac1, RhoA, TC10 and TCL were expressed in human corneal epithelial cells. Among them, Cdc42 and TCL were found to significantly affect cell migration in monolayer scratch assays. These results were confirmed through the use of validated siRNAs directed to Cdc42 and TCL. Scramble siRNA transfected cells had high percentage of polarized cells than Cdc42 or TCL siRNA transfected cells at the wound edge. We showed that the Cdc42-specific effector p21-activated kinase 4 localized predominantly to cell-cell junctions in cell monolayers, but failed to translocate to the leading edge in Cdc42 siRNA transfected cells after monolayer wounding. CONCLUSION:Rho proteins expressed in cultured human corneal epithelial cells, and Cdc42, TCL facilitate two-dimensional cell migration in-vitro. Although silencing of Cdc42 and TCL did not noticeably affect the appearance of cell adhesions at the leading edge, the slower migration of these cells indicates both GTP-binding proteins play important roles in promoting cell movement of human corneal epithelial cells.
- Published
- 2013
- Full Text
- View/download PDF
4. PTB domain-directed substrate targeting in a tyrosine kinase from the unicellular choanoflagellate Monosiga brevicollis.
- Author
-
Victoria Prieto-Echagüe, Perry M Chan, Barbara P Craddock, Edward Manser, and W Todd Miller
- Subjects
Medicine ,Science - Abstract
Choanoflagellates are considered to be the closest living unicellular relatives of metazoans. The genome of the choanoflagellate Monosiga brevicollis contains a surprisingly high number and diversity of tyrosine kinases, tyrosine phosphatases, and phosphotyrosine-binding domains. Many of the tyrosine kinases possess combinations of domains that have not been observed in any multicellular organism. The role of these protein interaction domains in M. brevicollis kinase signaling is not clear. Here, we have carried out a biochemical characterization of Monosiga HMTK1, a protein containing a putative PTB domain linked to a tyrosine kinase catalytic domain. We cloned, expressed, and purified HMTK1, and we demonstrated that it possesses tyrosine kinase activity. We used immobilized peptide arrays to define a preferred ligand for the third PTB domain of HMTK1. Peptide sequences containing this ligand sequence are phosphorylated efficiently by recombinant HMTK1, suggesting that the PTB domain of HMTK1 has a role in substrate recognition analogous to the SH2 and SH3 domains of mammalian Src family kinases. We suggest that the substrate recruitment function of the noncatalytic domains of tyrosine kinases arose before their roles in autoinhibition.
- Published
- 2011
- Full Text
- View/download PDF
5. Supplementary Figure 1 from PRL-3 Down-regulates PTEN Expression and Signals through PI3K to Promote Epithelial-Mesenchymal Transition
- Author
-
Qi Zeng, Jing Ping Tang, Edward Manser, Jing Ming Dong, Samantha Yiling Quah, and Haihe Wang
- Abstract
Supplementary Figure 1 from PRL-3 Down-regulates PTEN Expression and Signals through PI3K to Promote Epithelial-Mesenchymal Transition
- Published
- 2023
6. Tensin3 interaction with talin drives the formation of fibronectin-associated fibrillar adhesions
- Author
-
Paul Atherton, Rafaella Konstantinou, Suat Peng Neo, Emily Wang, Eleonora Balloi, Marina Ptushkina, Hayley Bennett, Kath Clark, Jayantha Gunaratne, David Critchley, Igor Barsukov, Edward Manser, and Christoph Ballestrem
- Subjects
Talin ,Focal Adhesions ,Integrins ,Tensins ,Cell Adhesion ,Cell Biology ,Vinculin ,Extracellular Matrix ,Fibronectins - Abstract
The formation of healthy tissue involves continuous remodeling of the extracellular matrix (ECM). Whilst it is known that this requires integrin-associated cell-ECM adhesion sites (CMAs) and actomyosin-mediated forces, the underlying mechanisms remain unclear. Here, we examine how tensin3 contributes to the formation of fibrillar adhesions (FBs) and fibronectin fibrillogenesis. Using BioID mass spectrometry and a mitochondrial targeting assay, we establish that tensin3 associates with the mechanosensors such as talin and vinculin. We show that the talin R11 rod domain binds directly to a helical motif within the central intrinsically disordered region (IDR) of tensin3, whilst vinculin binds indirectly to tensin3 via talin. Using CRISPR knock-out cells in combination with defined tensin3 mutations, we show (i) that tensin3 is critical for the formation of α5β1-integrin FBs and for fibronectin fibrillogenesis, and (ii) the talin/tensin3 interaction drives this process, with vinculin acting to potentiate it.
- Published
- 2022
7. Insights into the evolution of regulated actin dynamics via characterization of primitive gelsolin/cofilin proteins from Asgard archaea
- Author
-
Edward Manser, Robert Robinson, Linh T. Tran, Yohendran Baskaran, Laurent Blanchoin, Magali Orhant-Prioux, Caner Akıl, Agency for science, technology and research [Singapore] (A*STAR), Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Research Institute for Interdisciplinary Science, Okayama University, CytoMorphoLab, Physiologie cellulaire et végétale (LPCV), Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes (UGA)-Institut de Recherche Interdisciplinaire de Grenoble (IRIG), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes (UGA)-Institut de Recherche Interdisciplinaire de Grenoble (IRIG), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Vidyasirimedhi Institute of Science and Technology, Agency for Science, Technology and Research, Singapore National Medical Research Council (NMRC Grant OFIRG/0067/2018), Vidyasirimedhi Institute of Science and Technology, Research Institute for Interdisciplinary Science, Japan Society for the Promotion of Science (KAKENHI Grant JP20H00476), European Project: 741773,AAA, Department of Pharmacology, Yong Loo Lin School of Medicine [Singapour], National University of Singapore (NUS), Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche Interdisciplinaire de Grenoble (IRIG), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Université Grenoble Alpes (UGA)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche Interdisciplinaire de Grenoble (IRIG), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Université Grenoble Alpes (UGA), and Vidyasirimedhi Institute of Science and Technology [Thaïlande] (VISTEC)
- Subjects
Protein Conformation, alpha-Helical ,gelsolin ,Archaeal Proteins ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,macromolecular substances ,Biochemistry ,Polymerization ,Evolution, Molecular ,03 medical and health sciences ,0302 clinical medicine ,Genome, Archaeal ,Lokiarchaeota ,Amino Acid Sequence ,Cytoskeleton ,Actin ,X-ray crystallography ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,Actin filament severing ,Biological Sciences ,eukaryogenesis ,Cofilin ,musculoskeletal system ,Actin cytoskeleton ,Archaea ,Actins ,Cell biology ,Actin Depolymerizing Factors ,Asgard archaea ,Profilin ,biology.protein ,Sequence Alignment ,actin ,Filopodia ,Gelsolin ,030217 neurology & neurosurgery - Abstract
Significance Eukaryotic gelsolin superfamily proteins generally comprise three or more related domains. Here we characterize single- and double-domain gelsolins from Thorarchaeota (Thor). Similar domain architectures are present in Heimdall-, Loki-, and Odinarchaeota. Thor gelsolins are functional in regulating rabbit actin in in vitro assays, showing a range of activities including actin filament severing and bundling. These gelsolins bind to the eukaryotic gelsolin/cofilin-binding site on actin. Two-domain, but not one-domain, gelsolins are calcium regulated. Thor gelsolins appear to have the characteristics and structure consistent with primitive gelsolins/cofilins, suggesting that these single- and double-domain gelsolins are a record of a nascent preeukaryotic actin-regulation machinery., Asgard archaea genomes contain potential eukaryotic-like genes that provide intriguing insight for the evolution of eukaryotes. The eukaryotic actin polymerization/depolymerization cycle is critical for providing force and structure in many processes, including membrane remodeling. In general, Asgard genomes encode two classes of actin-regulating proteins from sequence analysis, profilins and gelsolins. Asgard profilins were demonstrated to regulate actin filament nucleation. Here, we identify actin filament severing, capping, annealing and bundling, and monomer sequestration activities by gelsolin proteins from Thorarchaeota (Thor), which complete a eukaryotic-like actin depolymerization cycle, and indicate complex actin cytoskeleton regulation in Asgard organisms. Thor gelsolins have homologs in other Asgard archaea and comprise one or two copies of the prototypical gelsolin domain. This appears to be a record of an initial preeukaryotic gene duplication event, since eukaryotic gelsolins are generally comprise three to six domains. X-ray structures of these proteins in complex with mammalian actin revealed similar interactions to the first domain of human gelsolin or cofilin with actin. Asgard two-domain, but not one-domain, gelsolins contain calcium-binding sites, which is manifested in calcium-controlled activities. Expression of two-domain gelsolins in mammalian cells enhanced actin filament disassembly on ionomycin-triggered calcium release. This functional demonstration, at the cellular level, provides evidence for a calcium-controlled Asgard actin cytoskeleton, indicating that the calcium-regulated actin cytoskeleton predates eukaryotes. In eukaryotes, dynamic bundled actin filaments are responsible for shaping filopodia and microvilli. By correlation, we hypothesize that the formation of the protrusions observed from Lokiarchaeota cell bodies may involve the gelsolin-regulated actin structures.
- Published
- 2020
8. Tensin3 interaction with talin drives formation of fibronectin-associated fibrillar adhesions
- Author
-
Igor L. Barsukov, Eleonora Balloi, David R. Critchley, Rafaella Konstantinou, Suat Peng Neo, Emily C.S. Wang, Hayley M. Bennett, Kath Clark, Jayantha Gunaratne, Paul Atherton, Marina Ptushkina, Edward Manser, and Christoph Ballestrem
- Subjects
Extracellular matrix ,Fibronectin ,animal structures ,biology ,Chemistry ,Mitochondrial targeting ,biology.protein ,Healthy tissue ,Fibrillogenesis ,macromolecular substances ,Adhesion ,Vinculin ,Cell biology - Abstract
The formation of healthy tissue involves continuous remodelling of the extracellular matrix (ECM). Whilst it is known that this requires integrin-associated cell-ECM adhesion sites (CMAs) and actomyosin-mediated forces, the underlying mechanisms remain unclear. Here we examine how tensin3 contributes to formation of fibrillar adhesions (FBs) and fibronectin fibrillo-genesis. Using BioID mass spectrometry and a mitochondrial targeting assay, we establish that tensin3 associates with the mechanosensors talin and vinculin. We show that the talin R11 rod domain binds directly to a helical motif within the central intrinsically disordered region (IDR) of tensin3, whilst vinculin binds indirectly to tensin3 via talin. Using CRISPR knock-out cells in combination with defined tensin3 mutations, we show (i) that tensin3 is critical for formation of α5β1-integrin FBs and for fibronectin fibrillogenesis, and (ii) the talin/tensin3 interaction drives this process, with vinculin acting to potentiate it.
- Published
- 2021
9. The structure of the actin filament uncapping complex mediated by twinfilin
- Author
-
Edward Manser, Robert Robinson, and Dennis M. Mwangangi
- Subjects
0303 health sciences ,Multidisciplinary ,Chemistry ,SciAdv r-articles ,macromolecular substances ,Biochemistry ,Protein–protein interaction ,Protein filament ,03 medical and health sciences ,0302 clinical medicine ,Biophysics ,030217 neurology & neurosurgery ,Uncapping ,Actin ,Research Articles ,030304 developmental biology ,Research Article - Abstract
Competitive interactions between capping protein, twinfilin, and actin drive actin filament uncapping and recycling., Uncapping of actin filaments is essential for driving polymerization and depolymerization dynamics from capping protein–associated filaments; however, the mechanisms of uncapping leading to rapid disassembly are unknown. Here, we elucidated the x-ray crystal structure of the actin/twinfilin/capping protein complex to address the mechanisms of twinfilin uncapping of actin filaments. The twinfilin/capping protein complex binds to two G-actin subunits in an orientation that resembles the actin filament barbed end. This suggests an unanticipated mechanism by which twinfilin disrupts the stable capping of actin filaments by inducing a G-actin conformation in the two terminal actin subunits. Furthermore, twinfilin disorders critical actin-capping protein interactions, which will assist in the dissociation of capping protein, and may promote filament uncapping through a second mechanism involving V-1 competition for an actin-binding surface on capping protein. The extensive interactions with capping protein indicate that the evolutionary conserved role of twinfilin is to uncap actin filaments.
- Published
- 2021
10. Proximity proteomics identifies PAK4 as a component of Afadin-Nectin junctions
- Author
-
Claire Lee Foon Swa, Edward Manser, Elsa Yuen Wai Ng, Sheena Wee, Jayantha Gunaratne, Felicia P. L. Tay, and Yohendran Baskaran
- Subjects
Proteomics ,Cell signaling ,Science ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Nectins ,General Physics and Astronomy ,Biotin ,CDC42 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mass Spectrometry ,Article ,Nectin ,Cell Line, Tumor ,Cell polarity ,Protein Interaction Mapping ,Humans ,Protein Isoforms ,Carbon-Nitrogen Ligases ,cdc42 GTP-Binding Protein ,Multidisciplinary ,Osteoblasts ,Kinase ,Cadherin ,Escherichia coli Proteins ,Gene Expression Profiling ,Microfilament Proteins ,General Chemistry ,Cell biology ,Repressor Proteins ,Intercellular Junctions ,Gene Expression Regulation ,p21-Activated Kinases ,Isotope Labeling ,Phosphorylation ,Protein Binding ,Signal Transduction ,Cell signalling - Abstract
Human PAK4 is an ubiquitously expressed p21-activated kinase which acts downstream of Cdc42. Since PAK4 is enriched in cell-cell junctions, we probed the local protein environment around the kinase with a view to understanding its location and substrates. We report that U2OS cells expressing PAK4-BirA-GFP identify a subset of 27 PAK4-proximal proteins that are primarily cell-cell junction components. Afadin/AF6 showed the highest relative biotin labelling and links to the nectin family of homophilic junctional proteins. Reciprocally >50% of the PAK4-proximal proteins were identified by Afadin BioID. Co-precipitation experiments failed to identify junctional proteins, emphasizing the advantage of the BioID method. Mechanistically PAK4 depended on Afadin for its junctional localization, which is similar to the situation in Drosophila. A highly ranked PAK4-proximal protein LZTS2 was immuno-localized with Afadin at cell-cell junctions. Though PAK4 and Cdc42 are junctional, BioID analysis did not yield conventional cadherins, indicating their spatial segregation. To identify cellular PAK4 substrates we then assessed rapid changes (12’) in phospho-proteome after treatment with two PAK inhibitors. Among the PAK4-proximal junctional proteins seventeen PAK4 sites were identified. We anticipate mammalian group II PAKs are selective for the Afadin/nectin sub-compartment, with a demonstrably distinct localization from tight and cadherin junctions., PAK4 is a kinase involved in cell-cell junctions, though the identify of the local protein network involving PAK4 is unclear. Here, the authors performed proximity proteomic analysis on mammalian PAK4 and find that PAK4 is associated with Afadin-dependent junctions, and report putative PAK4 phosphorylation substrates at this site.
- Published
- 2020
11. Complex eukaryotic-like actin regulation systems from Asgard archaea
- Author
-
Robert Robinson, Laurent Blanchoin, Magali Orhant-Prioux, Edward Manser, Linh T. Tran, Yohendran Baskaran, and Caner Akıl
- Subjects
Protein filament ,biology ,Chemistry ,Lokiarchaeota ,macromolecular substances ,Actin filament severing ,Actin cytoskeleton ,biology.organism_classification ,Filopodia ,Gelsolin ,Actin ,Cell biology ,Archaea - Abstract
Asgard archaea genomes contain potential eukaryotic-like genes that provide intriguing insight for the evolution of eukaryotes. The actin polymerization/depolymerization cycle is critical for providing force and structure for a variety of processes in eukaryotes, including membrane remodelling. Here, we identify actin filament severing, capping, annealing and bundling, and monomer sequestration activities by gelsolin proteins from Thorarchaeota (Thor), which complete a eukaryote-like actin depolymerization cycle. Thor gelsolins are comprised of one or two copies of the prototypical gelsolin domain and appear to be a record of an initial pre-eukaryotic gene duplication event, since eukaryotic gelsolins are generally comprised of three to six domains. X-ray crystal structure determination of these proteins in complex with mammalian actin revealed similar interactions to the first domain of human gelsolin. Asgard two-domain, but not one-domain, gelsolins contain calcium-binding sites, which is manifested in calcium-controlled activities. Expression of two-domain gelsolins in mammalian cells led to enhanced actin filament disassembly on ionomycin-triggered calcium release. This functional demonstration, at the cellular level, provides evidence for calcium-regulated actin cytoskeleton in Asgard archaea, and indicates that the calcium-regulated actin cytoskeleton predates eukaryotes. In eukaryotes, dynamic bundled filaments are responsible for shaping filopodia and microvilli. By correlation, the formation of the protrusions observed from Lokiarchaeota cell bodies may involve gelsolin-regulated actin structures.
- Published
- 2019
12. Group-I PAKs-mediated phosphorylation of HACE1 at serine 385 regulates its oligomerization state and Rac1 ubiquitination
- Author
-
Jérôme Boudeau, Amel Mettouchi, Serge Urbach, Orane Visvikis, Maria I. Acosta, Anne Doye, Emmanuel Lemichez, Yuen-Wai Ng, Anne Debant, Edward Manser, Centre méditerranéen de médecine moléculaire (C3M), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Génomique Fonctionnelle (IGF), Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS), Institute of Molecular and Cell Biology (IMCB), Epithelial Cell Biology Laboratory, Centre de recherche en Biologie Cellulaire (CRBM), Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 1 (UM1), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Côte d'Azur (UCA), and Université Montpellier 1 (UM1)-Université Montpellier 2 - Sciences et Techniques (UM2)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Proteomics ,Vascular Endothelial Growth Factor A ,rac1 GTP-Binding Protein ,Ubiquitin-Protein Ligases ,Mutant ,Bacterial Toxins ,lcsh:Medicine ,RAC1 ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Article ,Cell Line ,Serine ,03 medical and health sciences ,Ubiquitin ,Human Umbilical Vein Endothelial Cells ,Humans ,Phosphorylation ,p21-activated kinases ,lcsh:Science ,ComputingMilieux_MISCELLANEOUS ,Multidisciplinary ,biology ,Chemistry ,Escherichia coli Proteins ,lcsh:R ,Ubiquitination ,Cell biology ,Ubiquitin ligase ,Cytosol ,030104 developmental biology ,p21-Activated Kinases ,biology.protein ,lcsh:Q ,Protein Multimerization - Abstract
The regulation of Rac1 by HACE1-mediated ubiquitination and proteasomal degradation is emerging as an essential element in the maintenance of cell homeostasis. However, how the E3 ubiquitin ligase activity of HACE1 is regulated remains undetermined. Using a proteomic approach, we identified serine 385 as a target of group-I PAK kinases downstream Rac1 activation by CNF1 toxin from pathogenic E. coli. Moreover, cell treatment with VEGF also promotes Ser-385 phosphorylation of HACE1. We have established in vitro that HACE1 is a direct target of PAK1 kinase activity. Mechanistically, we found that the phospho-mimetic mutant HACE1(S385E), as opposed to HACE1(S385A), displays a lower capacity to ubiquitinate Rac1 in cells. Concomitantly, phosphorylation of Ser-385 plays a pivotal role in controlling the oligomerization state of HACE1. Finally, Ser-385 phosphorylated form of HACE1 localizes in the cytosol away from its target Rac1. Together, our data point to a feedback inhibition of HACE1 ubiquitination activity on Rac1 by group-I PAK kinases.
- Published
- 2018
13. Myotonic dystrophy kinase-related Cdc42-binding kinases (MRCK), the ROCK-like effectors of Cdc42 and Rac1
- Author
-
Edward Manser and Zhuo-shen Zhao
- Subjects
rho-Associated Kinases ,Protein family ,Major effector proteins for the Rho GTPases: Review ,Effector ,Kinase ,Molecular Sequence Data ,RAC1 ,macromolecular substances ,Cell Biology ,CDC42 ,GTPase ,Biology ,medicine.disease ,Second Messenger Systems ,Biochemistry ,Myotonic dystrophy ,Myotonin-Protein Kinase ,Cell biology ,medicine ,Animals ,Humans ,Amino Acid Sequence ,cdc42 GTP-Binding Protein ,Cell adhesion - Abstract
Cdc42 is a member of the Rho GTPase protein family that plays key roles in local F-actin organization through a number of kinase and non-kinase effector proteins. The myotonic dystrophy kinase-related Cdc42-binding kinases (MRCKs), and the RhoA binding coiled-coil containing kinases (ROCKs) are widely expressed members of the Dystrophia myotonica protein kinase (DMPK) family. The MRCK proteins are ∼190 kDa multi-domain proteins expressed in all cells and coordinate certain acto-myosin networks. Notably MRCK is a key regulator of myosin18A and myosin IIA/B, and through phosphorylation of their common regulatory light chains (MYL9 or MLC2) to promote actin stress fiber contractility. The MRCK kinases are regulated by Cdc42, which is required for cell polarity and directional migration; MRCK links to the acto-myosin complex through interaction with a coiled-coil containing adaptor proteins LRAP35a/b. The biological activities of MRCK in model organisms such as worms and flies confirm it as a myosin II activator. In mammalian cell culture MRCK can be critical for cancer cell migration and neurite outgrowth. We review the current literatures regarding MRCK and highlight the similarities and differences between MRCK and ROCK kinases.
- Published
- 2015
14. NMR binding and crystal structure reveal that intrinsically-unstructured regulatory domain auto-inhibits PAK4 by a mechanism different from that of PAK1
- Author
-
Jianxing Song, Liangzhong Lim, Wei Wang, Edward Manser, and Yohendran Baskaran
- Subjects
Magnetic Resonance Spectroscopy ,Protein Conformation ,Stereochemistry ,Molecular Sequence Data ,Group ii ,Biophysics ,Peptide ,Crystal structure ,CDC42 ,Biochemistry ,PAK1 ,Transferase ,Amino Acid Sequence ,Enzyme Inhibitors ,Protein Kinase Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,Crystallography ,Kinase ,Cell Biology ,Nuclear magnetic resonance spectroscopy ,p21-Activated Kinases ,chemistry ,Protein Binding - Abstract
Six human PAK members are classified into groups I (PAKs 1-3) and II (PAK4-6). Previously, only group I PAKs were thought to be auto-inhibited but very recently PAK4, the prototype of group II PAKs, has also been shown to be auto-inhibited by its N-terminal regulatory domain. However, the complete auto-inhibitory domain (AID) sequence remains undefined and the mechanism underlying its auto-inhibition is largely elusive. Here, the N-terminal regulatory domain of PAK4 sufficient for auto-inhibiting and binding Cdc42/Rac was characterized to be intrinsically unstructured, but nevertheless we identified the entire AID sequence by NMR. Strikingly, an AID peptide was derived by deleting the binding-unnecessary residues, which has a Kd of 320 nM to the PAK4 catalytic domain. Consequently, the PAK4 crystal structure complexed with the entire AID has been determined, which reveals that the complete kinase cleft is occupied by 20 AID residuescomposed of an N-terminal α-helix and a previously-identified pseudosubstrate motif, thus achieving auto-inhibition. Our study reveals that PAK4 is auto-inhibited by a novel mechanism which is completely different from that for PAK1, thus bearing critical implications for design of inhibitors specific for group II PAKs.
- Published
- 2013
15. Formin DAAM1 Organizes Actin Filaments in the Cytoplasmic Nodal Actin Network
- Author
-
Michael P. Sheetz, Edward Manser, Zi Zhao Lieu, Weiwei Luo, and Alexander D. Bershadsky
- Subjects
0301 basic medicine ,rho GTP-Binding Proteins ,Cytoplasm ,Cell Membranes ,lcsh:Medicine ,Arp2/3 complex ,Actin Filaments ,Microfilament ,Biochemistry ,Actin remodeling of neurons ,Mice ,0302 clinical medicine ,Contractile Proteins ,Macromolecular Structure Analysis ,lcsh:Science ,Microscopy ,Multidisciplinary ,biology ,Chemistry ,Microfilament Proteins ,Light Microscopy ,Cell biology ,Cell Motility ,Actin Cytoskeleton ,Protein Transport ,Cell Processes ,Lamellipodium ,Cellular Structures and Organelles ,Research Article ,Protein Structure ,Fluorescence Recovery after Photobleaching ,macromolecular substances ,Research and Analysis Methods ,Transfection ,03 medical and health sciences ,Animals ,Humans ,Actin-binding protein ,Molecular Biology Techniques ,Protein Structure, Quaternary ,Molecular Biology ,Actin ,Adaptor Proteins, Signal Transducing ,lcsh:R ,Cell Membrane ,Actin remodeling ,Biology and Life Sciences ,Proteins ,Cell Biology ,Actin cytoskeleton ,Actins ,Cytoskeletal Proteins ,030104 developmental biology ,biology.protein ,lcsh:Q ,MDia1 ,Protein Multimerization ,030217 neurology & neurosurgery ,Actin Polymerization ,HeLa Cells - Abstract
A nodal cytoplasmic actin network underlies actin cytoplasm cohesion in the absence of stress fibers. We previously described such a network that forms upon Latrunculin A (LatA) treatment, in which formin DAAM1 was localized at these nodes. Knock down of DAAM1 reduced the mobility of actin nodes but the nodes remained. Here we have investigated DAAM1 containing nodes after LatA washout. DAAM1 was found to be distributed between the cytoplasm and the plasma membrane. The membrane binding likely occurs through an interaction with lipid rafts, but is not required for F-actin assembly. Interesting the forced interaction of DAAM1 with plasma membrane through a rapamycin-dependent linkage, enhanced F-actin assembly at the cell membrane (compared to the cytoplasm) after the LatA washout. However, immediately after addition of both rapamycin and LatA, the cytoplasmic actin nodes formed transiently, before DAAM1 moved to the membrane. This was consistent with the idea that DAAM1 was initially anchored to cytoplasmic actin nodes. Further, photoactivatable tracking of DAAM1 showed DAAM1 was immobilized at these actin nodes. Thus, we suggest that DAAM1 organizes actin filaments into a nodal complex, and such nodal complexes seed actin network recovery after actin depolymerization.
- Published
- 2016
16. Proximity biotinylation provides insight into the molecular composition of focal adhesions at the nanometer scale
- Author
-
Thomas Leung, Felicia P. L. Tay, Hannah L. F. Swa, Jayantha Gunaratne, Jing-Ming Dong, Edward Manser, and Brian Burke
- Subjects
0301 basic medicine ,Role of cell adhesions in neural development ,Integrin ,Biochemistry ,Focal adhesion ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Chlorocebus aethiops ,Animals ,Humans ,Biotinylation ,Molecular Biology ,Paxillin ,Adaptor Proteins, Signal Transducing ,Focal Adhesions ,biology ,Chemistry ,Adhesome ,Tumor Suppressor Proteins ,Membrane Proteins ,Signal transducing adaptor protein ,Cell Biology ,Transmembrane protein ,Neoplasm Proteins ,Cell biology ,Cytoskeletal Proteins ,030104 developmental biology ,Membrane protein ,COS Cells ,biology.protein ,Apoptosis Regulatory Proteins ,Carrier Proteins ,Cell Adhesion Molecules ,030217 neurology & neurosurgery - Abstract
Focal adhesions are protein complexes that link metazoan cells to the extracellular matrix through the integrin family of transmembrane proteins. Integrins recruit many proteins to these complexes, referred to as the "adhesome." We used proximity-dependent biotinylation (BioID) in U2OS osteosarcoma cells to label proteins within 15 to 25 nm of paxillin, a cytoplasmic focal adhesion protein, and kindlin-2, which directly binds β integrins. Using mass spectrometry analysis of the biotinylated proteins, we identified 27 known adhesome proteins and 8 previously unknown components close to paxillin. However, only seven of these proteins interacted directly with paxillin, one of which was the adaptor protein Kank2. The proteins in proximity to β integrin included 15 of the adhesion proteins identified in the paxillin BioID data set. BioID also correctly established kindlin-2 as a cell-cell junction protein. By focusing on this smaller data set, new partners for kindlin-2 were found, namely, the endocytosis-promoting proteins liprin β1 and EFR3A, but, contrary to previous reports, not the filamin-binding protein migfilin. A model adhesome based on both data sets suggests that focal adhesions contain fewer components than previously suspected and that paxillin lies away from the plasma membrane. These data not only illustrate the power of using BioID and stable isotope-labeled mass spectrometry to define macromolecular complexes but also enable the correct identification of therapeutic targets within the adhesome.
- Published
- 2016
17. Group I and II mammalian PAKs have different modes of activation by Cdc42
- Author
-
Widyawilis Selamat, Felicia Tay Pei Ling, Edward Manser, Yuen-Wai Ng, and Yohendran Baskaran
- Subjects
Molecular Sequence Data ,macromolecular substances ,CDC42 ,environment and public health ,Biochemistry ,Group (periodic table) ,Chlorocebus aethiops ,Serine ,Genetics ,Animals ,Amino Acid Sequence ,Phosphorylation ,cdc42 GTP-Binding Protein ,Cytoskeleton ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,biology ,Effector ,Kinase ,Scientific Reports ,biology.organism_classification ,Amino acid ,Enzyme Activation ,enzymes and coenzymes (carbohydrates) ,p21-Activated Kinases ,chemistry ,COS Cells ,Mutation ,Protozoa ,biological phenomena, cell phenomena, and immunity - Abstract
p21-activated kinases (PAKs) are Cdc42 effectors found in metazoans, fungi and protozoa. They are subdivided into PAK1-like (group I) or PAK4-like (group II) kinases. Human PAK4 is widely expressed and its regulatory mechanism is unknown. We show that PAK4 is strongly inhibited by a newly identified auto-inhibitory domain (AID) formed by amino acids 20 to 68, which is evolutionarily related to that of other PAKs. In contrast to group I kinases, PAK4 is constitutively phosphorylated on Ser 474 in the activation loop, but held in an inactive state until Cdc42 binding. Thus, group II PAKs are regulated through conformational changes in the AID rather than A-loop phosphorylation.
- Published
- 2012
18. The PAKs come of age
- Author
-
Edward Manser and Jeffrey Field
- Subjects
Cell signaling ,Effector ,Kinase ,Focal adhesion assembly ,macromolecular substances ,Cell Biology ,GTPase ,CDC42 ,Biology ,Bioinformatics ,environment and public health ,Biochemistry ,enzymes and coenzymes (carbohydrates) ,PAK1 ,parasitic diseases ,Synaptic plasticity ,Molecular Medicine ,biological phenomena, cell phenomena, and immunity ,Neuroscience - Abstract
Protein kinases are versatile signaling molecules that are involved in the regulation most physiological responses. The p21-activated kinases (PAKs) can be activated directly by the small GTPases Rac and Cdc42 and are among the best characterized downstream effectors of these Rho proteins. The structure, substrate specificity and functional role of PAKS are evolutionarily conserved from protozoa to mammals. Vertebrate PAKs are particularly important for cytoskeletal remodeling and focal adhesion assembly, thereby contributing to dynamic processes such as cell migration and synaptic plasticity. This issue of Cellular Logistics focuses on the PAK family of kinases, with ten reviews written by researchers currently working in the field. Here in this introductory overview we highlight some of the most interesting recent discoveries regarding PAK biochemistry and biology. The reviews in this issue cover a range of topics including the atomic structures of PAK1 and PAK4, their role in animals as assessed by knockout studies, and how PAKs are likely to contribute to cancer and neurodegenerative diseases. The promise remains that PAK inhibitors will emerge that validate current pre-clinical studies suggesting that blocking PAK activity will positively contribute to human health.
- Published
- 2012
19. Regulation of IRSp53-Dependent Filopodial Dynamics by Antagonism between 14-3-3 Binding and SH3-Mediated Localization
- Author
-
Edward Manser, Lee Yeow-Fong, Elsa Ng, Christine Hall, and Jeffrey Robens
- Subjects
animal structures ,Dendritic spine morphogenesis ,Nerve Tissue Proteins ,macromolecular substances ,CDC42 ,Biology ,environment and public health ,SH3 domain ,src Homology Domains ,EPS8 ,Cell Movement ,Chlorocebus aethiops ,Animals ,Humans ,Pseudopodia ,Phosphorylation ,cdc42 GTP-Binding Protein ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Binding Sites ,Intracellular Signaling Peptides and Proteins ,Signal transducing adaptor protein ,Articles ,Cell Biology ,Actin cytoskeleton ,Wiskott-Aldrich Syndrome Protein Family ,Cell biology ,14-3-3 Proteins ,COS Cells ,Mutation ,Lamellipodium ,Filopodia ,HeLa Cells ,Protein Binding - Abstract
Filopodia are dynamic structures found at the leading edges of most migrating cells. IRSp53 plays a role in filopodium dynamics by coupling actin elongation with membrane protrusion. IRSp53 is a Cdc42 effector protein that contains an N-terminal inverse-BAR (Bin-amphipysin-Rvs) domain (IRSp53/MIM homology domain [IMD]) and an internal SH3 domain that associates with actin regulatory proteins, including Eps8. We demonstrate that the SH3 domain functions to localize IRSp53 to lamellipodia and that IRSp53 mutated in its SH3 domain fails to induce filopodia. Through SH3 domain-swapping experiments, we show that the related IRTKS SH3 domain is not functional in lamellipodial localization. IRSp53 binds to 14-3-3 after phosphorylation in a region that lies between the CRIB and SH3 domains. This association inhibits binding of the IRSp53 SH3 domain to proteins such as WAVE2 and Eps8 and also prevents Cdc42-GTP interaction. The antagonism is achieved by phosphorylation of two related 14-3-3 binding sites at T340 and T360. In the absence of phosphorylation at these sites, filopodium lifetimes in cells expressing exogenous IRSp53 are extended. Our work does not conform to current views that the inverse-BAR domain or Cdc42 controls IRSp53 localization but provides an alternative model of how IRSp53 is recruited (and released) to carry out its functions at lamellipodia and filopodia.
- Published
- 2010
20. Cytoplasmic ACK1 Interaction with Multiple Receptor Tyrosine Kinases Is Mediated by Grb2
- Author
-
Lin Pao-Chun, Perry M. Chan, Edward Manser, and Wing Chan
- Subjects
AXL receptor tyrosine kinase ,biology ,GAS6 ,Cell Biology ,Biochemistry ,Tropomyosin receptor kinase C ,Receptor tyrosine kinase ,Cell biology ,ROR1 ,Cancer research ,biology.protein ,Anaplastic lymphoma kinase ,GRB2 ,Molecular Biology ,Tyrosine kinase - Abstract
ACK1 (activated Cdc42-associated kinase 1), a cytoplsmic tyrosine kinase, is implicated in metastatic behavior, cell spreading and migration, and epidermal growth factor receptor (EGFR) signaling. The function of ACK1 in the regulation of receptor tyrosine kinases requires a C-terminal region that demonstrates a significant homology to the EGFR binding domain of MIG6. In this study, we have identified additional receptor tyrosine kinases, including Axl, leukocyte tyrosine kinase, and anaplastic lymphoma kinase, that can bind to the ACK1/MIG6 homology region. Unlike the interaction between MIG6 and EGFR, our data suggest that these receptor tyrosine kinases require the adaptor protein Grb2 for efficient binding, which interacts with highly conserved proline-rich regions that are conserved between ACK1 and MIG6. We have focused on Axl and compared how ACK1/Axl differs from the ACK1/EGFR axis by investigating effects of knockdown of endogenous ACK1. Although EGFR activation promotes ACK1 turnover, Axl activation by GAS6 does not; interestingly, the reciprocal down-regulation of GAS6-stimulated Axl is blocked by removing ACK1. Thus, ACK1 functions in part to control Axl receptor levels. Silencing of ACK1 also leads to diminished ruffling and migration in DU145 and COS7 cells upon GAS6-Axl signaling. The ability of ACK1 to modulate Axl and perhaps anaplastic lymphoma kinase (altered in anaplastic large cell lymphomas) might explain why ACK1 can promote metastatic and transformed behavior in a number of cancers.
- Published
- 2009
21. Stat3 promotes directional cell migration by regulating Rac1 activity via its activator βPIX
- Author
-
Terk Shin Teng, Xinmin Cao, Dominic C.H. Ng, Edward Manser, and Baohong Lin
- Subjects
STAT3 Transcription Factor ,Transcriptional Activation ,rac1 GTP-Binding Protein ,Cytoplasm ,RAC1 ,Gene Knockout Techniques ,Mice ,Cell Movement ,Stress Fibers ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,STAT3 ,Cells, Cultured ,Actin ,biology ,Activator (genetics) ,Cell growth ,Cell migration ,Cell Biology ,Fibroblasts ,Actin cytoskeleton ,Actins ,Cell biology ,Gene Knockdown Techniques ,biology.protein ,STAT protein ,Rho Guanine Nucleotide Exchange Factors - Abstract
Stat3 is a member of the signal transducer and activator of transcription family, which is important for cytokine signaling as well as for a number of cellular processes including cell proliferation, anti-apoptosis and immune responses. In recent years, evidence has emerged suggesting that Stat3 also participates in cell invasion and motility. However, how Stat3 regulates these processes remains poorly understood. Here, we find that loss of Stat3 expression in mouse embryonic fibroblasts leads to an elevation of Rac1 activity, which promotes a random mode of migration by reducing directional persistence and formation of actin stress fibers. Through rescue experiments, we demonstrate that Stat3 can regulate the activation of Rac1 to mediate persistent directional migration and that this function is not dependent on Stat3 transcriptional activity. We find that Stat3 binds to βPIX, a Rac1 activator, and that this interaction could represent a mechanism by which cytoplasmic Stat3 regulates Rac1 activity to modulate the organization of actin cytoskeleton and directional migration.
- Published
- 2009
22. Paxillin nuclear-cytoplasmic localization is regulated by phosphorylation of the LD4 motif: evidence that nuclear paxillin promotes cell proliferation
- Author
-
Edward Manser, Louis Lim, Yuen-Wai Ng, Jing-Ming Dong, and Lei-Shong Lau
- Subjects
Cytoplasm ,Amino Acid Motifs ,PTK2 ,Active Transport, Cell Nucleus ,Biochemistry ,Cell Line ,Zyxin ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Phosphorylation ,Nuclear export signal ,Molecular Biology ,Paxillin ,Cell Proliferation ,LIM domain ,Cell Nucleus ,Focal Adhesions ,biology ,Cell Biology ,Molecular biology ,Cell nucleus ,medicine.anatomical_structure ,Gene Expression Regulation ,Fatty Acids, Unsaturated ,biology.protein ,Signal transduction - Abstract
Paxillin, a major focal-adhesion complex component belongs to the subfamily of LIM domain proteins and participates in cell adhesion-mediated signal transduction. It is implicated in cell-motility responses upon activation of cell-surface receptors and can recruit, among others, the GIT1 [GRK (G-protein-coupled-receptor kinase)-interacting ARF (ADP-ribosylation factor) GAP (GTPase-activating protein)]–PIX [PAK (p21-activated kinase)-interacting exchange factor]–PAK1 complex. Several adhesion proteins including zyxin, Hic5 and Trip6 are also nuclear and can exert transcriptional effects. In the present study we show that endogenous paxillin shuttles between the cytoplasm and nucleus, and we have used a variety of tagged paxillin constructs to map the nuclear export signal. This region overlaps an important LD4 motif that binds GIT1 and FAK1 (focal-adhesion kinase 1). We provide evidence that phosphorylation of Ser272 within LD4 blocks nuclear export, and we show that this modification also reduces GIT1, but not FAK1, binding; however, Ser272 phosphorylation does not appear to be mediated by PAK1 as previously suggested. Expression of nuclear-localized paxillin LIM domains stimulate DNA synthesis and cell proliferation. By real-time PCR analysis we have established that overexpression of either full-length paxillin or a truncated nuclear form suppresses expression of the parental imprinted gene H19, and modulation of this locus probably affects the rate of NIH-3T3 cell proliferation.
- Published
- 2009
23. The Drosophila homologue of Arf-GAP GIT1, dGIT, is required for proper muscle morphogenesis and guidance during embryogenesis
- Author
-
Louis Lim, Xiaohang Yang, Juliana M. Choy, Edward Manser, and Sami M. Bahri
- Subjects
Cell signaling ,Embryo, Nonmammalian ,GTPase-activating protein ,Muscle Fibers, Skeletal ,Mutant ,Morphogenesis ,Embryonic Development ,Penetrance ,Biology ,Transfection ,Muscle guidance ,dGIT ,Cell Adhesion ,Muscle attachment ,Animals ,Drosophila Proteins ,Wings, Animal ,Molecular Biology ,Alleles ,dPak ,Sequence Homology, Amino Acid ,dPIX ,Myogenesis ,Muscles ,GTPase-Activating Proteins ,Cell Biology ,Cell biology ,Protein Transport ,Drosophila melanogaster ,Phenotype ,p21-Activated Kinases ,Mutation ,Adhesion ,Drosophila ,Axon guidance ,Developmental biology ,Developmental Biology - Abstract
GIT1-like proteins are GTPase-activating proteins (GAPs) for Arfs and interact with a variety of signaling molecules to function as integrators of pathways controlling cytoskeletal organization and cell motility. In this report, we describe the characterization of a Drosophila homologue of GIT1, dGIT, and show that it is required for proper muscle morphogenesis and myotube guidance in the fly embryo. The dGIT protein is concentrated at the termini of growing myotubes and localizes to muscle attachment sites in late stage embryos. dgit mutant embryos show muscle patterning defects and aberrant targeting in subsets of their muscles. dgit mutant muscles fail to localize the p21-activated kinase, dPak, to their termini. dPak and dGIT form a complex in the presence of dPIX and dpak mutant embryos show similar muscle morphogenesis and targeting phenotypes to that of dgit. We propose that dGIT and dPak are part of a complex that promotes proper muscle morphogenesis and myotube targeting during embryogenesis.
- Published
- 2009
24. PAK Is Regulated by PI3K, PIX, CDC42, and PP2Cα and Mediates Focal Adhesion Turnover in the Hyperosmotic Stress-induced p38 Pathway
- Author
-
Louis Lim, Perry M. Chan, and Edward Manser
- Subjects
MAPK/ERK pathway ,MAP Kinase Signaling System ,p38 mitogen-activated protein kinases ,macromolecular substances ,Biology ,p38 Mitogen-Activated Protein Kinases ,environment and public health ,Biochemistry ,Focal adhesion ,Phosphatidylinositol 3-Kinases ,PAK1 ,Osmotic Pressure ,Chlorocebus aethiops ,parasitic diseases ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Protein Phosphatase 2 ,cdc42 GTP-Binding Protein ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Focal Adhesions ,Activator (genetics) ,Kinase ,Mechanisms of Signal Transduction ,PTEN Phosphohydrolase ,Cell Biology ,Molecular biology ,Protein Structure, Tertiary ,Rats ,Enzyme Activation ,enzymes and coenzymes (carbohydrates) ,p21-Activated Kinases ,COS Cells ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Rho Guanine Nucleotide Exchange Factors ,HeLa Cells - Abstract
Fractionation of brain extracts and functional biochemical assays identified PP2Calpha, a serine/threonine phosphatase, as the major biochemical activity inhibiting PAK1. PP2Calpha dephosphorylated PAK1 and p38, both of which were activated upon hyperosmotic shock with the same kinetics. In comparison to growth factors, hyperosmolality was a more potent activator of PAK1. Therefore we characterize the PAK signaling pathway in the hyperosmotic shock response. Endogenous PAKs were recruited to the p38 kinase complex in a phosphorylation-dependent manner. Overexpression of a PAK inhibitory peptide or dominant negative Cdc42 revealed that p38 activation was dependent on PAK and Cdc42 activities. PAK mutants deficient in binding to Cdc42 or PAK-interacting exchange factor were not activated. Using a panel of kinase inhibitors, we identified PI3K acting upstream of PAK, which correlated with PAK repression by pTEN overexpression. RNA interference knockdown of PAK expression reduced stress-induced p38 activation and conversely, PP2Calpha knockdown increased its activation. Hyperosmotic stress-induced PAK translocation away from focal adhesions to the perinuclear compartment and resulted in disassembly of focal adhesions, which are hallmarks of PAK activation. Inhibition of PAK by overexpression of PP2Calpha or the kinase inhibitory domain prevented sorbitol-induced focal adhesion dissolution. Inhibition of MAPK pathways showed that MEK-ERK signaling but not p38 is required for full PAK activation and focal adhesion turnover. We conclude that 1) PAK plays a required role in hyperosmotic signaling through the PI3K/pTEN/Cdc42/PP2Calpha/p38 pathway, and 2) PAK and PP2Calpha modulate the effects of this pathway on focal adhesion dynamics.
- Published
- 2008
25. Autophosphorylation-dependent degradation of Pak1, triggered by the Rho-family GTPase, Chp
- Author
-
Natalia Volinsky, Edward Manser, Ami Aronheim, Monika Hubsman, and Deborah Yablonski
- Subjects
rho GTP-Binding Proteins ,Proteasome Endopeptidase Complex ,T-Lymphocytes ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Biology ,Biochemistry ,SH3 domain ,Cell Line ,PAK1 ,Cell Movement ,GTP-Binding Proteins ,Animals ,Humans ,Phosphorylation ,Kinase activity ,Protein kinase A ,Molecular Biology ,Ubiquitin ,Kinase ,Autophosphorylation ,Cell Biology ,Neoplasm Proteins ,Gene Expression Regulation ,p21-Activated Kinases ,Tyrosine kinase ,Protein Binding ,Signal Transduction ,Research Article - Abstract
The Paks (p21-activated kinases) Pak1, Pak2 and Pak3 are among the most studied effectors of the Rho-family GTPases, Rac, Cdc42 (cell division cycle 42) and Chp (Cdc42 homologous protein). Pak kinases influence a variety of cellular functions, but the process of Pak down-regulation, following activation, is poorly understood. In the present study, we describe for the first time a negative-inhibitory loop generated by the small Rho-GTPases Cdc42 and Chp, resulting in Pak1 inhibition. Upon overexpression of Chp, we unexpectedly observed a T-cell migration phenotype consistent with Paks inhibition. In line with this observation, overexpression of either Chp or Cdc42 caused a marked reduction in the level of Pak1 protein in a number of different cell lines. Chp-induced degradation was accompanied by ubiquitination of Pak1, and was dependent on the proteasome. The susceptibility of Pak1 to Chp-induced degradation depended on its p21-binding domain, kinase activity and a number of Pak1 autophosphorylation sites, whereas the PIX- (Pak-interacting exchange factor) and Nck-binding sites were not required. Together, these results implicate Chp-induced kinase autophosphorylation in the degradation of Pak1. The N-terminal domain of Chp was found to be required for Chp-induced degradation, although not for Pak1 activation, suggesting that Chp provides a second function, distinct from kinase activation, to trigger Pak degradation. Collectively, our results demonstrate a novel mechanism of signal termination mediated by the Rho-family GTPases Chp and Cdc42, which results in ubiquitin-mediated degradation of one of their direct effectors, Pak1. Abbreviations: Cdc42, cell division cycle 42; Chp, Cdc42 homologous protein; DTT, dithiothreitol; EGFP, enhanced green fluorescent protein; ERK, extracellular-signal-regulated kinase; HA, haemagglutinin; HEK-293, human embryonic kidney; Hrs, HGF (hepatocyte growth factor)-regulated tyrosine kinase substrate; KI, kinase inhibitory; MAPK, mitogen-activated protein kinase; MBP, myelin basic protein; NP-40, Nonidet P40; Pak, p21-activated kinase; Paki, Pak inhibitor; PBD, p21-binding domain; PI, protease inhibitor; PIX, Pak-interacting exchange factor; POSH, plenty of SH3s; SDF-1α, stromal-cell derived factor-1α; SH3, Src homology 3 domain; tTA, tet-transactivator; WCE, whole-cell extract
- Published
- 2007
26. PRL-3 Down-regulates PTEN Expression and Signals through PI3K to Promote Epithelial-Mesenchymal Transition
- Author
-
Samantha Yiling Quah, Edward Manser, Jing Ping Tang, Qi Zeng, Haihe Wang, and Jing Ming Dong
- Subjects
rac1 GTP-Binding Protein ,endocrine system ,Cancer Research ,endocrine system diseases ,Integrin ,Down-Regulation ,CHO Cells ,Transfection ,Mesoderm ,Focal adhesion ,Phosphatidylinositol 3-Kinases ,Cricetulus ,Cricetinae ,Animals ,Humans ,PTEN ,Tensin ,Epithelial–mesenchymal transition ,Phosphorylation ,PI3K/AKT/mTOR pathway ,Paxillin ,biology ,PTEN Phosphohydrolase ,Epithelial Cells ,Actins ,Vinculin ,Neoplasm Proteins ,Oncology ,Cancer cell ,Cancer research ,biology.protein ,Protein Tyrosine Phosphatases ,Colorectal Neoplasms ,rhoA GTP-Binding Protein ,hormones, hormone substitutes, and hormone antagonists ,HeLa Cells ,Signal Transduction - Abstract
PRL-3 is a metastasis-associated phosphatase. We and others have shown that its overexpression increases cell motility and invasiveness. These phenotypic changes are reminiscent of the epithelial-mesenchymal transition (EMT) that occurs during embryonic development and oncogenesis. The EMT is a complex process that converts epithelia into migratory mesenchymal cells. We here attempt to unravel the underlying mechanistic basis of these phenomena. HeLa cells transiently expressing EGFP-PRL-3 (HeLa-PRL-3) exhibit reduced levels of paxillin. Similarly, Chinese hamster ovary cells stably expressing myc-PRL-3 (CHO-PRL-3) also show marked reductions in paxillin, phosphorylated paxillin-Tyr31, and vinculin at focal adhesion complexes and notable reductions in the levels of RhoA-GTP, Rac1-GTP, and filamentous-actin filaments. DLD-1 human colorectal cancer cells engineered to express EGFP-PRL-3 (DLD-1-PRL-3) underwent changes consistent with EMT. In these cells, PRL-3 activates Akt and inactivates glycogen synthase kinase-3β as assessed by phosphospecific antibodies. PRL-3 up-regulates mesenchymal markers fibronectin and Snail and down-regulates epithelial markers E-cadherin, γ-catenin (plakoglobin), and integrin β3, which are major effectors in the EMT pathway. The changes in these EMT characteristics brought about by PRL-3 can be abrogated by the phosphoinositide 3-kinase (PI3K) inhibitor LY294002, implying that PRL-3 acts upstream of PI3K and could play an initiating role to trigger the EMT switch during cancer metastasis. In addition, PRL-3 can down-regulate phosphatase and tensin homologue deleted on chromosome 10, which is an important antagonist of PI3K, further reinforcing PI3K/Akt function in PRL-3–triggered EMT. Catalytically inactive PRL-3 (C104S) was impaired in the above PRL-3–mediated events, indicating that these properties require phosphatase activity. Targeting PRL-3 may thus be a useful strategy to impede cancer cell invasion and metastasis. [Cancer Res 2007;67(7):2922–6]
- Published
- 2007
27. An in cellulo-derived structure of PAK4 in complex with its inhibitor Inka1
- Author
-
Edward Manser, Khay C. Ang, Robert Robinson, Jonathan M. Grimes, Wee L. Chan, Praju V. Anekal, and Yohendran Baskaran
- Subjects
Immunoprecipitation ,General Physics and Astronomy ,Crystal structure ,Plasma protein binding ,In Vitro Techniques ,Biology ,Crystallography, X-Ray ,Article ,General Biochemistry, Genetics and Molecular Biology ,Crystal ,chemistry.chemical_compound ,Adenosine Triphosphate ,Catalytic Domain ,Cell Line, Tumor ,Chlorocebus aethiops ,Escherichia coli ,Animals ,Humans ,Microscopy, Confocal ,Multidisciplinary ,COS cells ,Kinase ,HEK 293 cells ,Intracellular Signaling Peptides and Proteins ,General Chemistry ,Protein Structure, Tertiary ,3. Good health ,HEK293 Cells ,p21-Activated Kinases ,Biochemistry ,chemistry ,COS Cells ,Biophysics ,Crystallization ,Adenosine triphosphate ,HeLa Cells ,Protein Binding - Abstract
PAK4 is a metazoan-specific kinase acting downstream of Cdc42. Here we describe the structure of human PAK4 in complex with Inka1, a potent endogenous kinase inhibitor. Using single mammalian cells containing crystals 50 μm in length, we have determined the in cellulo crystal structure at 2.95 Å resolution, which reveals the details of how the PAK4 catalytic domain binds cellular ATP and the Inka1 inhibitor. The crystal lattice consists only of PAK4–PAK4 contacts, which form a hexagonal array with channels of 80 Å in diameter that run the length of the crystal. The crystal accommodates a variety of other proteins when fused to the kinase inhibitor. Inka1–GFP was used to monitor the process crystal formation in living cells. Similar derivatives of Inka1 will allow us to study the effects of PAK4 inhibition in cells and model organisms, to allow better validation of therapeutic agents targeting PAK4., PAK4 is a metazoan-specific kinase, which acts downstream of the cell polarity regulator Cdc42. Here, Baskaran et al. determine the structure of PAK4 bound to the endogenous inhibitor Inka1 from crystals that form spontaneously in mammalian cells overexpressing both proteins.
- Published
- 2015
28. The GIT-Associated Kinase PAK Targets to the Centrosome and Regulates Aurora-A
- Author
-
Louis Lim, Edward Manser, Jet Phey Lim, Yuen Wai Ng, and Zhuo-shen Zhao
- Subjects
Molecular Sequence Data ,Mitosis ,Cell Cycle Proteins ,macromolecular substances ,Biology ,Protein Serine-Threonine Kinases ,environment and public health ,Focal adhesion ,Histones ,Mice ,PAK1 ,Aurora Kinases ,Chlorocebus aethiops ,Animals ,Humans ,Protein Isoforms ,Phosphorylation ,RNA, Small Interfering ,Molecular Biology ,Aurora Kinase A ,Centrosome ,Kinase ,GTPase-Activating Proteins ,Cell Biology ,Phosphoproteins ,Spindle apparatus ,Cell biology ,Rats ,enzymes and coenzymes (carbohydrates) ,p21-Activated Kinases ,COS Cells ,NIH 3T3 Cells ,biological phenomena, cell phenomena, and immunity ,HeLa Cells - Abstract
Previously, we showed PAK-PIX-GIT targets and regulates focal adhesions; here, we uncover a different function for the complex at the centrosome. Active PAK1 is particularly evident in mitosis and phosphorylates the centrosomal adaptor GIT1 on serine 517. Interestingly, direct centrosome targeting activates the kinase via a process not requiring Rho GTPases; excision of the centrosome prevents this activation. Once activated, PAK1 dissociates from PIX/GIT but can bind to and phosphorylate the important centrosomal kinase Aurora-A. PAK1 promotes phosphorylation of Aurora-A on Thr288 and Ser342, which are key sites for kinase activation in mitosis. In vivo PAK activation causes an accumulation of activated Aurora-A; conversely, when betaPIX is depleted or PAK is inhibited, there is a delay in centrosome maturation. These observations may underlie reported effects of active PAK on cells, including histone H3 phosphorylation, alterations in centrosome number, and progression through mitosis.
- Published
- 2005
- Full Text
- View/download PDF
29. SNX9 as an adaptor for linking synaptojanin-1 to the Cdc42 effector ACK1
- Author
-
Louis Lim, Lee Yeow-Fong, and Edward Manser
- Subjects
Recombinant Fusion Proteins ,Molecular Sequence Data ,Vesicular Transport Proteins ,Biophysics ,Biotin ,Nerve Tissue Proteins ,Signalling ,Plasma protein binding ,Synaptojanin ,Biology ,Biochemistry ,Clathrin ,src Homology Domains ,Structural Biology ,Genetics ,Animals ,Drosophila Proteins ,Humans ,Amino Acid Sequence ,Cdc42 ,cdc42 GTP-Binding Protein ,Sorting Nexins ,Molecular Biology ,Src homology domain ,Base Sequence ,ACK ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,Protein-Tyrosine Kinases ,Phosphoric Monoester Hydrolases ,Synaptojanin-1 ,Sorting nexin ,Cdc42 GTP-Binding Protein ,biology.protein ,Carrier Proteins ,SNX9 ,Protein Binding ,Binding domain - Abstract
Sorting nexin 9 (SNX9, also referred to as SH3PX1) is a binding partner for the non-receptor and Cdc42-associated kinase (ACK) in Drosophila and mammals. ACK1 is known to bind clathrin and influence EGF receptor endocytosis. SNX9 comprises an N-terminal Src homology domain 3 (SH3), a central PHOX homology (PX) domain, and a carboxyl-terminal coiled-coil region. In order to investigate SNX9 further we have made use of a novel in vivo biotinylation system to label various GST-SH3 domains and perform blot overlays, thereby identifying synaptojanin-1 as a partner for SNX9. Biotinylated SH3 domains were also used for specific identification of target proline-rich sequences in synaptojanin and ACK1 on synthetic peptides arrays. Direct assessment of SH3 binding efficiencies at different positions within the extensive proline-rich regions of these proteins were thus determined. While SNX9 targets a number of sequences within the proline-rich regions of synaptojanin, a single site was identified in human ACK1. By testing the association of various truncations of ACK1 with SNX9 we confirmed the dominant SNX9 binding domain in human ACK1 (residues 920–955). In the presence of SNX9 we find that synaptojanin is able to colocalize with distinct ACK1 containing vesicles, indicating that this tyrosine kinase is linked to many components involved in vesicle dynamics including clathrin, AP2 and synaptojanin-1.
- Published
- 2005
30. GIT1 Activates p21-Activated Kinase through a Mechanism Independent of p21 Binding
- Author
-
Tsui-Han Loo, Edward Manser, Louis Lim, and Yuen-Wai Ng
- Subjects
rac1 GTP-Binding Protein ,GTPase-activating protein ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Cell Cycle Proteins ,macromolecular substances ,Protein Serine-Threonine Kinases ,Biology ,environment and public health ,Focal adhesion ,Mice ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Amino Acid Sequence ,cdc42 GTP-Binding Protein ,p21-activated kinases ,Cell Growth and Development ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Focal Adhesions ,Kinase ,GTPase-Activating Proteins ,Autophosphorylation ,Signal transducing adaptor protein ,Cell Biology ,Phosphoproteins ,Protein Structure, Tertiary ,Cell biology ,Enzyme Activation ,enzymes and coenzymes (carbohydrates) ,p21-Activated Kinases ,Cdc42 GTP-Binding Protein ,Mutation ,NIH 3T3 Cells ,Guanine nucleotide exchange factor ,biological phenomena, cell phenomena, and immunity ,Rho Guanine Nucleotide Exchange Factors ,HeLa Cells ,Protein Binding ,Subcellular Fractions - Abstract
p21-activated kinases (PAKs) associate with a guanine nucleotide exchange factor, Pak-interacting exchange factor (PIX), which in turn binds the paxillin-associated adaptor GIT1 that targets the complex to focal adhesions. Here, a detailed structure-function analysis of GIT1 reveals how this multidomain adaptor also participates in activation of PAK. Kinase activation does not occur via Cdc42 or Rac1 GTPase binding to PAK. The ability of GIT1 to stimulate alphaPAK autophosphorylation requires the participation of the GIT N-terminal Arf-GAP domain but not Arf-GAP activity and involves phosphorylation of PAK at residues common to Cdc42-mediated activation. Thus, the activation of PAK at adhesion complexes involves a complex interplay between the kinase, Rho GTPases and protein partners that provide localization cues.
- Published
- 2004
31. Cdc42-dependent nuclear translocation of non-receptor tyrosine kinase, ACK
- Author
-
Edward Manser, Yolanda Calle, Ijaz Ahmed, Alam Nur-E-Kamal, Mohammed A Sayed, Jabeen M Kamal, Sally Meiners, and Padmanabhan Rengaswamy
- Subjects
rho GTP-Binding Proteins ,Non-receptor tyrosine kinase ,Cytoplasm ,Time Factors ,Transcription, Genetic ,GTPase ,PC12 Cells ,Biochemistry ,Mice ,Cytosol ,cdc42 GTP-Binding Protein ,Cytoskeleton ,Genetics ,Reverse Transcriptase Polymerase Chain Reaction ,Brain ,Protein-Tyrosine Kinases ,Immunohistochemistry ,Cell biology ,Electrophoresis, Polyacrylamide Gel ,Signal transduction ,Tyrosine kinase ,Cell Division ,Plasmids ,Protein Binding ,Signal Transduction ,Blotting, Western ,Molecular Sequence Data ,Active Transport, Cell Nucleus ,Biophysics ,macromolecular substances ,Biology ,Transfection ,Models, Biological ,Cell Line ,Cell Line, Tumor ,Animals ,Humans ,Amino Acid Sequence ,Nuclear export signal ,Molecular Biology ,Cell Nucleus ,Wound Healing ,Sequence Homology, Amino Acid ,Cell Biology ,Actin cytoskeleton ,Actins ,Protein Structure, Tertiary ,Rats ,Nuclear localization sequence - Abstract
Ras signals for the transformation of mammalian cells are apparently transduced through Rho GTPases. The Rho GTPase family member Cdc42 generates independent signals that regulate the rearrangement of the actin cytoskeleton and the transcription of genes. However, the molecular mechanism of signal transduction from Cdc42 to the nucleus remains to be understood. The non-receptor tyrosine kinases ACK-1 and ACK-2 have been found to bind specifically to Cdc42. In this paper we studied whether ACKs transduce Cdc42 signals to the nucleus directly, or through other cytoplasmic proteins. Using immunocytochemistry and Western blot analysis, we found a nuclear localization of ACKs in semi-confluent glioblastoma (U251) cells, as opposed to a cytosolic localization in confluent cells. In agreement with the nuclear localization, a putative nuclear export signal was identified in ACK-1 and ACK-2. Furthermore, the interaction of Cdc42 with ACKs was shown to be essential for the nuclear localization of ACKs. Overexpression of ACK42 (a Cdc42 binding domain of ACK) inhibited cell growth and movement, indicating that Cdc42 signals are transduced to the nucleus through ACKs. This is the first report providing evidence of a novel role for ACKs in transducing Cdc42 signals directly to the nucleus.
- Published
- 2004
32. PDZK1: I. A major scaffolder in brush borders of proximal tubular cells11See Editorial by Moe, p. 1916
- Author
-
Edward Manser, Zhuo-shen Zhao, Andrea Gantenbein, Akira Tsuji, Sandra Pribanic, Heini Murer, Jürg Biber, Patrik Forrer, Desa Bacic, Luc A. Sabourin, and Serge M. Gisler
- Subjects
NaPi-IIa ,yeast two-hybrid ,Two-hybrid screening ,PDZ domain ,renal transport of phosphate ,03 medical and health sciences ,0302 clinical medicine ,NaPi-I ,030304 developmental biology ,0303 health sciences ,Organic cation transport proteins ,biology ,PDZ proteins ,Apical membrane ,CFEX ,Cell biology ,Solute carrier family ,MAP17 ,Sodium–hydrogen antiporter ,Biochemistry ,Membrane protein ,Nephrology ,PDZK1 ,biology.protein ,URAT1 ,Cotransporter ,NHERF-1 ,D-AKAP2 ,030217 neurology & neurosurgery - Abstract
PDZK1: I. A major scaffolder in brush borders of proximal tubular cells. Background In proximal tubular cells, PDZK1 (NaPi-Cap1) has been implicated in apical expression of the Na+-dependent phosphate cotransporter (NaPi-IIa) via interaction with its C-terminus. PDZK1 represents a multidomain protein consisting of four PDZ domains and thus is believed to have a broader specificity besides NaPi-IIa. Methods We subjected single PDZ domains derived from PDZK1 either to yeast two-hybrid screens or yeast trap assays. Different pull-down assays and blot overlays were applied to corroborate the PDZK1-mediated interactions in vitro. Co-localization of interacting proteins with PDZK1 in proximal tubular cells was assessed by immunohistochemistry. Results In the yeast screens, the most abundant candidate protein to interact with PDZK1 was the membrane-associated protein of 17 kD (MAP17). Besides MAP17, C-terminal parts of following transporters were also identified: NaPi-IIa, solute carrier SLC17A1 (NaPi-I), Na+/H+ exchanger (NHE-3), organic cation transporter (OCTN1), chloride-formate exchanger (CFEX), and urate-anion exchanger (URAT1). In addition, other regulatory factors were found among the clones, such as a protein kinase A (PKA)-anchoring protein (D-AKAP2) and N+/H+ exchanger regulator factor (NHERF-1). All interactions of itemized proteins with PDZK1 were affirmed by in vitro techniques. Apart from PDZK1, strong in vitro interactions of NHERF-1 were also observed with the solute transporters (excluding MAP17) and D-AKAP2. All identified proteins were immunolocalized in proximal tubular cells, wherein all membrane proteins co-localized with PDZK1 in brush borders. Conclusion We hypothesize that PDZK1 and NHERF-1 establish an extended network beneath the apical membrane to which membrane proteins and regulatory components are anchored.
- Published
- 2003
- Full Text
- View/download PDF
33. Small GTPases Take the Stage
- Author
-
Edward Manser
- Subjects
GTPase-activating protein ,Upstream and downstream (transduction) ,GTPase-Activating Proteins ,Cell Biology ,Computational biology ,GTPase ,Biology ,Microtubules ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Eukaryotic Cells ,GTP-binding protein regulators ,Microtubule ,Ran ,Animals ,Humans ,Signal transduction ,Molecular Biology ,Eukaryotic cell ,Monomeric GTP-Binding Proteins ,Signal Transduction ,Developmental Biology - Abstract
Small GTPases are molecular switches that have been adopted to control many eukaryotic cell functions. Starting with the study of the protooncogene Ras in the early 1980s, detailed pathways have been uncovered upstream and downstream of Ras-related GTP binding proteins. Nonetheless, novel members have been discovered at a pace that has outstripped cell biologists, and thus much remains to be established regarding newer family members. Undiscovered functions are still being uncovered for “established” small GTPases such as Ras, Rho, and Ran. The topics covered at this meeting indeed demonstrate that Ras proteins are at the heart of cellular dynamics.
- Published
- 2002
- Full Text
- View/download PDF
34. Cdc42 antagonizes inductive action of cAMP on cell shape, via effects of the myotonic dystrophy kinase-related Cdc42-binding kinase (MRCK) on myosin light chain phosphorylation
- Author
-
Thomas Leung, Louis Lim, Edward Manser, and Jing-Ming Dong
- Subjects
Botulinum Toxins ,Myosin Light Chains ,Histology ,RHOA ,Myosin light-chain kinase ,Microinjections ,Recombinant Fusion Proteins ,macromolecular substances ,CDC42 ,Protein Serine-Threonine Kinases ,Cell morphology ,Cell Line ,Pathology and Forensic Medicine ,Focal adhesion ,Myosin ,Cyclic AMP ,Animals ,Humans ,Phosphorylation ,cdc42 GTP-Binding Protein ,Myosin-Light-Chain Kinase ,Cell Size ,ADP Ribose Transferases ,Focal Adhesions ,biology ,Kinase ,Colforsin ,Cell Biology ,General Medicine ,Immunohistochemistry ,Cell biology ,Mutation ,Second messenger system ,biology.protein ,rhoA GTP-Binding Protein - Abstract
Rho GTPases play pivotal roles in regulating cell morphology. We previously showed that RhoA acts via ROKalpha to counteract the effects of the classical second messenger cyclic AMP on cell shape changes. Here we show that active Cdc42V12 also competes against the cAMP-induced stellate morphology in SH-EP cells. This Cdc42 effect is not mediated by the RhoA/ ROK pathway but rather the related MRCKalpha, a myotonic dystrophy kinase-related Cdc42-binding kinase. Co-expression of a dominant inhibitory MRCKalpha mutant with Cdc42V12 blocks the ability of the GTPase to counteract cAMP, suggesting that MRCK acts downstream of Cdc42 in this process. Cdc42V12 enhances the phosphorylation of myosin light chain (MLC) at the cell periphery and sustains focal adhesion complexes, while MLC kinase inhibitors destroy focal adhesion complexes and impair the Cdc42V12 protective effect. The data suggest that the maintenance of focal adhesion complexes via the regulation of myosin II activity underlies the ability of Cdc42 to protect against the effect of elevated cAMP.
- Published
- 2002
35. Phosphorylation and reorganization of vimentin by p21-activated kinase (PAK)
- Author
-
Hidemasa Goto, Masaki Inagaki, Louis Lim, Yoshihiro Yasui, Edward Manser, and Kazushi Tanabe
- Subjects
Neurofilament ,biology ,Kinase ,Vimentin ,macromolecular substances ,Cell Biology ,environment and public health ,Molecular biology ,Cell biology ,Lim kinase ,enzymes and coenzymes (carbohydrates) ,Genetics ,biology.protein ,Phosphorylation ,Cytoskeleton ,Intermediate filament ,Actin - Abstract
Background: Intermediate filament (IF) is one of the three major cytoskeletal filaments. Vimentin is the most widely expressed IF protein component. The Rho family of small GTPases, such as Cdc42, Rac and Rho, are thought to control the organization of actin filaments as well as other cytoskeletal filaments. Results: We determined if the vimentin filaments can be regulated by p21-activated kinase (PAK), one of targets downstream of Cdc42 or Rac. In vitro analyses revealed that vimentin served as an excellent substrate for PAK. This phosphorylated vimentin lost the potential to form 10 nm filaments. We identified Ser25, Ser38, Ser50, Ser65 and Ser72 in the amino-terminal head domain as the major phosphorylation sites on vimentin for PAK. The ectopic expression of constitutively active PAK in COS-7 cells induced vimentin phosphorylation. Fibre bundles or granulates of vimentin were frequent in these transfected cells. However, the kinase-inactive mutant induced neither vimentin phosphorylation nor filament reorganization. Conclusion: Our observations suggest that PAK may regulate the reorganization of vimentin filaments through direct vimentin phosphorylation.
- Published
- 2002
36. Arg kinase-binding protein 2 (ArgBP2) interaction with α-actinin and actin stress fibers inhibits cell migration
- Author
-
Edward Manser, Jeffery Yong, and Praju Vikas Anekal
- Subjects
Cell division ,macromolecular substances ,Actinin ,Biology ,Transfection ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Cell Movement ,Cell Line, Tumor ,Chlorocebus aethiops ,Animals ,Humans ,Neoplasm Metastasis ,Phosphorylation ,Cytoskeleton ,Protein kinase A ,Molecular Biology ,Actin ,Adaptor Proteins, Signal Transducing ,Homeodomain Proteins ,COS cells ,Colforsin ,RNA-Binding Proteins ,Cell migration ,Cell Biology ,Actomyosin ,Actins ,Cell biology ,14-3-3 Proteins ,Microscopy, Fluorescence ,COS Cells ,Kinase binding ,HeLa Cells ,Protein Binding ,Signal Transduction - Abstract
Cell migration requires dynamic remodelling of the actomyosin network. We report here that an adapter protein, ArgBP2 is a component of α-actinin containing stress fibres and inhibits migration. ArgBP2 is undetectable in many commonly studied cancer derived cell lines. COS-7 and HeLa cells express ArgBP2 (by western analysis), but expression was detectable only in ~half the cells by immuno-fluorescence. Short-term clonal analysis demonstrated 0.2- 0.3% of cells switch ArgBP2 expression (on or off) per cell division. ArgBP2 can have a fundamental impact on the actomyosin network: ArgBP2 positive COS-7 cells, for example, are clearly distinguishable by their denser actomyosin (stress fibre) network. ArgBP2γ binding to α-actinin appears to underlie its ability to localize to stress fibres and to decrease cell migration. We map a small α-actinin binding region in ArgBP2 (residues 192-228) that is essential for these effects. Protein kinase A phosphorylation of ArgBP2γ at neighbouring Ser259 and consequent 14-3-3 binding blocks its interaction with α-actinin. ArgBP2 is known to be down-regulated in some aggressively metastatic cancers. Our work provides a biochemical explanation for the antimigratory effect of ArgBP2.
- Published
- 2014
37. β1PIX, the PAK-interacting exchange factor, requires localization via a coiled-coil region to promote microvillus-like structures and membrane ruffles
- Author
-
Zhou-shen Zhao, Cheng-Gee Koh, Louis Lim, Edward Manser, and Chee-Peng Ng
- Subjects
Recombinant Fusion Proteins ,Molecular Sequence Data ,Cell Cycle Proteins ,CDC42 ,GTPase ,Protein Serine-Threonine Kinases ,Biology ,Cell Fractionation ,Mice ,Structure-Activity Relationship ,Protein structure ,Chlorocebus aethiops ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Protein Isoforms ,Amino Acid Sequence ,Phosphorylation ,Coiled coil ,Binding Sites ,COS cells ,Microvilli ,Sequence Homology, Amino Acid ,C-terminus ,Cell Membrane ,3T3 Cells ,Cell Biology ,Microvillus ,Protein Structure, Tertiary ,Rats ,Cell biology ,medicine.anatomical_structure ,p21-Activated Kinases ,Biochemistry ,COS Cells ,Microscopy, Electron, Scanning ,Guanine nucleotide exchange factor ,Dimerization ,Rho Guanine Nucleotide Exchange Factors ,HeLa Cells - Abstract
PIX is a Rho-family guanine nucleotide exchange factor that binds PAK. We previously described two isoforms of PIX that differ in their N termini. Here, we report the identification of a new splice variant of betaPIX, designated beta2PIX, that is the dominant species in brain and that lacks the region of approximately 120 residues with predicted coiled-coil structure at the C terminus of beta1PIX. Instead, beta2PIX contains a serine-rich C terminus. To determine whether these splice variants differ in their cellular function, we studied the effect of expressing these proteins in HeLa cells. We found that the coiled-coil region plays a key role in the localization of beta1PIX to the cell periphery and is also responsible for PIX dimerization. Overexpression of beta1, but not beta2PIX, drives formation of membrane ruffles and microvillus-like structures (via activation of Rac1 and Cdc42, respectively), indicating that its function requires localized activation of these GTPases. Thus, beta1PIX, like other RhoGEFs, exerts specific morphological functions that are dependent on its intracellular location and are mediated by its C-terminal dimerization domain.
- Published
- 2001
38. Blot Overlays with 32P-Labeled Fusion Proteins
- Author
-
Edward Manser, Louis Lim, and Zhou-shen Zhao
- Subjects
Oncogene Proteins, Fusion ,GTP' ,Blotting, Western ,Protozoan Proteins ,Biology ,Polymerase Chain Reaction ,DNA-binding protein ,General Biochemistry, Genetics and Molecular Biology ,SH3 domain ,src Homology Domains ,Radioligand Assay ,chemistry.chemical_compound ,Adenosine Triphosphate ,Animals ,Transferase ,Phosphorylation ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Glutathione Transferase ,Oncogene Proteins ,Binding Sites ,Glutathione ,Fusion protein ,Rats ,Blot ,chemistry ,Biochemistry ,Molecular Probes ,Electrophoresis, Polyacrylamide Gel ,Guanosine Triphosphate ,Phosphorus Radioisotopes - Abstract
Proteins labeled with 32 P can be used as sensitive "prime" in blot overlays to detect binding proteins or domains. Small G-protein Ras can bind GTP with extremely high affinity (K d ∼ 10 −11 - 10 −12 M) in the presence of Mg 2+ . We have taken advantage of this property of Ras to develop a vector that expresses proteins of interest such as glutathione S -transferase (GST)/Ras fusion proteins for noncovalent labeling with [γ- 32 P]GTP. The labeling efficiency of this method is >60% and involves a single short incubation step. We have previously identified several binding proteins for the second SH3 domain of the adaptor Nck using this method. Here we illustrate the overlay method using the GST/Ras system and compare results with the SH3 domain labeled by phosphorylation with [γ- 32 P]ATP. Both methods are similarly specific and sensitive; however, we show that signals are dependent primarily on GST-mediated probe dimerization. These dimeric probes allow a more stable probe-target complex similar to immunoglobulin interactions, thus significantly improving the sensitivity of the technique.
- Published
- 2001
39. The Mechanism of PAK Activation
- Author
-
Lydia Tan, Louis Lim, Claire Chong, and Edward Manser
- Subjects
Protein-Serine-Threonine Kinases ,Kinase ,Autophosphorylation ,macromolecular substances ,Cell Biology ,Biology ,environment and public health ,Biochemistry ,Cell biology ,Serine ,enzymes and coenzymes (carbohydrates) ,Cdc42 GTP-Binding Protein ,Phosphorylation ,p21-activated kinases ,Molecular Biology ,MAPK14 - Abstract
The p21-activated kinases (PAKs), in common with many kinases, undergo multiple autophosphorylation events upon interaction with appropriate activators. The Cdc42-induced phosphorylation of PAK serves in part to dissociate the kinase from its partners PIX and Nck. Here we investigate in detail how autophosphorylation events affect the catalytic activity of PAK by altering the autophosphorylation sites in both α- and βPAK. Bothin vivo and in vitro analyses demonstrate that, although most phosphorylation events in the PAK N-terminal regulatory domain play no direct role in activation, a phosphorylation of αPAK serine 144 or βPAK serine 139, which lie in the kinase inhibitory domain, significantly contribute to activation. By contrast, sphingosine-mediated activation is independent of this residue, indicating a different mode of activation. Thus two autophosphorylation sites direct activation while three others control association with focal complexes via PIX and Nck.
- Published
- 2001
40. The Tyrosine Kinase ACK1 Associates with Clathrin-coated Vesicles through a Binding Motif Shared by Arrestin and Other Adaptors
- Author
-
Edward Manser, Lydia Tan, Louis Lim, and Mabel Teo
- Subjects
Endocytosis ,Biochemistry ,Clathrin ,Adaptor Protein Complex alpha Subunits ,Mice ,Arrestin ,Animals ,Molecular Biology ,biology ,Vesicle ,Membrane Proteins ,Signal transducing adaptor protein ,Biological Transport ,Clathrin-Coated Vesicles ,3T3 Cells ,Cell Biology ,Protein-Tyrosine Kinases ,Cell biology ,Adaptor Proteins, Vesicular Transport ,COS Cells ,biology.protein ,Clathrin adaptor proteins ,Tyrosine kinase ,Signal Transduction - Abstract
One target for the small GTPase Cdc42 is the nonreceptor tyrosine kinase activated Cdc42-associated kinase (ACK), which binds selectively to Cdc42.GTP. We report that ACK1 can associate directly with the heavy chain of clathrin. A central region in ACK1 containing a conserved motif behaves as a clathrin adaptor and competes with beta-arrestin for a common binding site on the clathrin N-terminal head domain. Overexpressed ACK1 perturbs clathrin distribution, an activity dependent on the presence of C-terminal "adaptor" sequences that are also present in the related nonkinase gene 33. ACK1 interacts with the adaptor Nck via SH3 interactions but does not form a trimeric complex with p21-activated serine/threonine kinase, which also binds Nck. Stable low level expression of green fluorescent protein-ACK1 in NIH 3T3 cells has been used to localize ACK1 to clathrin-containing vesicles. The co-localization of ACK1 in vivo with clathrin and AP-2 indicates that it participates in trafficking, underlying an ability to increase receptor-mediated transferrin uptake.
- Published
- 2001
41. p21-Activated Kinase PAK Phosphorylates Desmin at Sites Different from Those for Rho-Associated Kinase
- Author
-
Edward Manser, Ichiro Izawa, Louis Lim, Waro Taki, Hiroyasu Inada, Kazuhiro Ohtakara, Hidemasa Goto, and Masaki Inagaki
- Subjects
rac1 GTP-Binding Protein ,Recombinant Fusion Proteins ,Intermediate Filaments ,Biophysics ,macromolecular substances ,Protein Serine-Threonine Kinases ,Peptide Mapping ,environment and public health ,Biochemistry ,Desmin ,Phosphoserine ,chemistry.chemical_compound ,Animals ,Phosphorylation ,cdc42 GTP-Binding Protein ,Intermediate filament ,p21-activated kinases ,Molecular Biology ,rho-Associated Kinases ,biology ,Kinase ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,musculoskeletal system ,Molecular biology ,Protein Structure, Tertiary ,Rats ,Enzyme Activation ,Molecular Weight ,Kinetics ,enzymes and coenzymes (carbohydrates) ,Caldesmon ,p21-Activated Kinases ,Cdc42 GTP-Binding Protein ,chemistry ,biology.protein ,Guanosine Triphosphate ,biological phenomena, cell phenomena, and immunity ,rhoA GTP-Binding Protein - Abstract
p21-activated kinase (PAK) and Rho-associated kinase (Rho-kinase) have been shown to induce Ca(2+)-independent contraction of smooth muscle. PAK-induced contraction of Triton-skinned smooth muscle correlates with increased phosphorylation of caldesmon and desmin, although the role of desmin phosphorylation has remained obscure. Here we report that desmin serves as an excellent substrate for PAK in vitro. PAK phosphorylated desmin in a GTP. Cdc42/Rac-dependent manner. Phosphorylation of desmin by PAK dramatically inhibited its filament-forming ability. PAK phosphorylated mainly serine residues of the head domain of desmin, and the major phosphorylation sites differed from those for Rho-kinase. These results suggest that different site-specific phosphorylation of desmin via two divergent protein kinases downstream of Rho family GTPases would seem to increase the regulatory potential for organization of desmin filaments.
- Published
- 2000
42. Coupling of PAK-Interacting Exchange Factor PIX to GIT1 Promotes Focal Complex Disassembly
- Author
-
Tsui-Han Loo, Louis Lim, Edward Manser, and Zhou-shen Zhao
- Subjects
DNA, Complementary ,Saccharomyces cerevisiae Proteins ,GTPase-activating protein ,Cell Cycle Proteins ,macromolecular substances ,Protein Serine-Threonine Kinases ,Transfection ,Models, Biological ,Focal adhesion ,Fungal Proteins ,src Homology Domains ,Cell Movement ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Microscopy, Phase-Contrast ,p21-activated kinases ,Cell Growth and Development ,Molecular Biology ,Paxillin ,Cytoskeleton ,Adaptor Proteins, Signal Transducing ,Glutathione Transferase ,Oncogene Proteins ,biology ,GTPase-Activating Proteins ,Signal transducing adaptor protein ,Epithelial Cells ,Cell Biology ,Fibroblasts ,Phosphoproteins ,Precipitin Tests ,Cell biology ,Protein Structure, Tertiary ,rac GTP-Binding Proteins ,Rac GTP-Binding Proteins ,Cytoskeletal Proteins ,p21-Activated Kinases ,COS Cells ,biology.protein ,Guanine nucleotide exchange factor ,Signal transduction ,Chickens ,Rho Guanine Nucleotide Exchange Factors ,HeLa Cells ,Plasmids ,Protein Binding ,Signal Transduction - Abstract
The p21-activated kinase PAK is targeted to focal complexes (FCs) through interactions with the SH3 domains of the PAK-interacting exchange factor PIX and Nck. PIX is a Rac GTP exchange factor that also binds the G-protein-coupled receptor kinase-interacting protein known as GIT1. Overexpression of GIT1 in fibroblasts or epithelial cells causes a loss of paxillin from FCs and stimulates cell motility. This is due to the direct interaction of a C-terminal 125-residue domain of GIT1 with paxillin, under the regulation of PIX. In its activated state, GIT1 can promote FC disassembly independent of actin-myosin contractile events. Additionally, GIT directly couples to a key component of FCs, focal adhesion kinase (FAK), via a conserved Spa2 homology domain. We propose that GIT1 and FAK cooperate to promote motility both by directly regulating focal complex dynamics and by the activation of Rac.
- Published
- 2000
43. Melanoma chondroitin sulphate proteoglycan regulates cell spreading through Cdc42, Ack-1 and p130cas
- Author
-
Edward Manser, James B. McCarthy, Melanie A. Simpson, Leo T. Furcht, Joji Iida, Louis Lim, Kathryn M. Eisenmann, Kouichi Tachibana, Patricia J. Keely, and Jun-Lin Guan
- Subjects
Integrins ,Receptors, Lymphocyte Homing ,macromolecular substances ,Integrin alpha4beta1 ,Biology ,Transfection ,chemistry.chemical_compound ,Cell Movement ,Tumor Cells, Cultured ,Humans ,Neoplasm Invasiveness ,Phosphorylation ,Phosphotyrosine ,cdc42 GTP-Binding Protein ,Chondroitin Sulfate Proteoglycan 4 ,Melanoma ,NG2 proteoglycan ,Retinoblastoma-Like Protein p130 ,Proteins ,Tyrosine phosphorylation ,Cell Biology ,Protein-Tyrosine Kinases ,Phosphoproteins ,Precipitin Tests ,Cell biology ,Enzyme Activation ,Crk-Associated Substrate Protein ,Chondroitin Sulfate Proteoglycans ,chemistry ,Cdc42 GTP-Binding Protein ,CSPG4 ,Mutation ,Cancer research ,Signal transduction ,Tyrosine kinase ,Protein Binding ,Signal Transduction - Abstract
Melanoma chondroitin sulphate proteoglycan (MCSP) is a cell-surface antigen that has been implicated in the growth and invasion of melanoma tumours. Although this antigen is expressed early in melanoma progression, its biological function is unknown. MCSP can stimulate the integrin-alpha4 beta1-mediated adhesion and spreading of melanoma cells. Here we show that stimulated MCSP recruits tyrosine-phosphorylated p130 cas, an adaptor protein important in tumour cell motility and invasion. MCSP stimulation also results in a pronounced activation and recruitment of the Rho-family GTPase Cdc42. MCSP-induced spreading of melanoma cells is dependent upon active Cdc42, a Cdc42-associated tyrosine kinase (Ack-1) and tyrosine phosphorylation of p130cas. Furthermore, vectors inhibiting Ack-1 or Cdc42 expression and/or function abrogate MCSP-induced tyrosine phosphorylation and recruitment of p130cas. Our findings indicate that MCSP may modify tumour growth or invasion by a unique signal-transduction pathway that links Cdc42 activation to downstream tyrosine phosphorylation and subsequent cytoskeletal reorganization.
- Published
- 1999
44. Structure of the small G protein Cdc42 bound to the GTPase-binding domain of ACK
- Author
-
Peter N. Lowe, Louis Lim, Helen R. Mott, Ernest D. Laue, Daniel Nietlispach, Edward Manser, and Darerca Owen
- Subjects
Models, Molecular ,Magnetic Resonance Spectroscopy ,GTPase-activating protein ,Protein Conformation ,G protein ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Cell Cycle Proteins ,Small G Protein ,macromolecular substances ,GTPase ,Biology ,GTP Phosphohydrolases ,GTP-binding protein regulators ,Protein structure ,GTP-Binding Proteins ,Escherichia coli ,Humans ,Amino Acid Sequence ,cdc42 GTP-Binding Protein ,Conserved Sequence ,Multidisciplinary ,Sequence Homology, Amino Acid ,Effector ,Protein-Tyrosine Kinases ,Cell biology ,Biochemistry ,Cdc42 GTP-Binding Protein ,Protein Binding - Abstract
The proteins Cdc42 and Rac are members of the Rho family of small GTPases (G proteins), which control signal-transduction pathways that lead to rearrangements of the cell cytoskeleton, cell differentiation and cell proliferation. They do so by binding to downstream effector proteins. Some of these, known as CRIB (for Cdc42/Rac interactive-binding) proteins, bind to both Cdc42 and Rac, such as the PAK1-3 serine/threonine kinases, whereas others are specific for Cdc42, such as the ACK tyrosine kinases and the Wiscott-Aldrich-syndrome proteins (WASPs). The effector loop of Cdc42 and Rac (comprising residues 30-40, also called switch I), is one of two regions which change conformation on exchange of GDP for GTP. This region is almost identical in Cdc42 and Racs, indicating that it does not determine the specificity of these G proteins. Here we report the solution structure of the complex of Cdc42 with the GTPase-binding domain ofACK. Both proteins undergo significant conformational changes on binding, to form a new type of G-protein/effector complex. The interaction extends the beta-sheet in Cdc42 by binding an extended strand from ACK, as seen in Ras/effector interactions, but it also involves other regions of the G protein that are important for determining the specificity of effector binding.
- Published
- 1999
45. PAK promotes morphological changes by acting upstream of Rac
- Author
-
Axel Obermeier, Louis Lim, Christine Hall, Sohail Ahmed, Su Ching Yen, and Edward Manser
- Subjects
Microinjections ,Membrane ruffling ,Genetic Vectors ,macromolecular substances ,GTPase ,Protein Serine-Threonine Kinases ,Biology ,PC12 Cells ,General Biochemistry, Genetics and Molecular Biology ,Substrate Specificity ,PAK1 ,GTP-binding protein regulators ,Cell Movement ,GTP-Binding Proteins ,Neurites ,Animals ,Chemical Precipitation ,Phosphorylation ,p21-activated kinases ,Molecular Biology ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,General Immunology and Microbiology ,General Neuroscience ,Cell Membrane ,JNK Mitogen-Activated Protein Kinases ,Cell Differentiation ,Molecular biology ,Guanine Nucleotides ,Rats ,rac GTP-Binding Proteins ,Cell biology ,Rac GTP-Binding Proteins ,p21-Activated Kinases ,Calcium-Calmodulin-Dependent Protein Kinases ,Mutagenesis, Site-Directed ,Guanine nucleotide exchange factor ,Mitogen-Activated Protein Kinases ,biological phenomena, cell phenomena, and immunity ,Lamellipodium ,Signal Transduction ,Subcellular Fractions ,Research Article - Abstract
The serine/threonine kinase p21-activated kinase (PAK) has been implicated as a downstream effector of the small GTPases Rac and Cdc42. While these GTPases evidently induce a variety of morphological changes, the role(s) of PAK remains elusive. Here we report that overexpression of betaPAK in PC12 cells induces a Rac phenotype, including cell spreading/membrane ruffling, and increased lamellipodia formation at growth cones and shafts of nerve growth factor-induced neurites. These effects are still observed in cells expressing kinase-negative or Rac/Cdc42 binding-deficient PAK mutants, indicating that kinase- and p21-binding domains are not involved. Furthermore, lamellipodia formation in all cell lines, including those expressing Rac binding-deficient PAK, is inhibited significantly by dominant-negative RacN17. Equal inhibition is achieved by blocking PAK interaction with the guanine nucleotide exchange factor PIX using a specific N-terminal PAK fragment. We conclude that PAK, via its N-terminal non-catalytic domain, acts upstream of Rac mediating lamellipodia formation through interaction with PIX.
- Published
- 1998
46. A Conserved Negative Regulatory Region in αPAK: Inhibition of PAK Kinases Reveals Their Morphological Roles Downstream of Cdc42 and Rac1
- Author
-
Thomas Leung, Edward Manser, Zhou-shen Zhao, Claire Chong, Xiang-Qun Chen, and Louis Lim
- Subjects
Protein Conformation ,Proto-Oncogene Proteins c-akt ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Cell Cycle Proteins ,macromolecular substances ,Protein Serine-Threonine Kinases ,Biology ,Focal adhesion ,GTP-binding protein regulators ,GTP-Binding Proteins ,Escherichia coli ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,cdc42 GTP-Binding Protein ,p21-activated kinases ,Cytoskeleton ,Cell Growth and Development ,Molecular Biology ,Binding Sites ,Protein-Serine-Threonine Kinases ,Kinase ,Cell Biology ,Molecular biology ,Actins ,Enzyme Activation ,Amino Acid Substitution ,Cdc42 GTP-Binding Protein ,COS Cells ,Mutagenesis, Site-Directed ,HeLa Cells ,Protein Binding - Abstract
AlphaPAK in a constitutively active form can exert morphological effects (E. Manser, H.-Y. Huang, T.-H. Loo, X.-Q. Chen, J.-M. Dong, T. Leung, and L. Lim, Mol. Cell. Biol. 17:1129-1143, 1997) resembling those of Cdc42G12V. PAK family kinases, conserved from yeasts to humans, are directly activated by Cdc42 or Rac1 through interaction with a conserved N-terminal motif (corresponding to residues 71 to 137 in alphaPAK). alphaPAK mutants with substitutions in this motif that resulted in severely reduced Cdc42 binding can be recruited normally to Cdc42G12V-driven focal complexes. Mutation of residues in the C-terminal portion of the motif (residues 101 to 137), though not affecting Cdc42 binding, produced a constitutively active kinase, suggesting this to be a negative regulatory region. Indeed, a 67-residue polypeptide encoding alphaPAK83-149 potently inhibited GTPgammaS-bound Cdc42-mediated kinase activation of both alphaPAK and betaPAK. Coexpression of this PAK inhibitor with Cdc42G12V prevented the formation of peripheral actin microspikes and associated loss of stress fibers normally induced by the p21. Coexpression of PAK inhibitor with Rac1G12V also prevented loss of stress fibers but not ruffling induced by the p21. Coexpression of alphaPAK83-149 completely blocked the phenotypic effects of hyperactive alphaPAKL107F in promoting dissolution of focal adhesions and actin stress fibers. These results, coupled with previous observations with constitutively active PAK, demonstrate that these kinases play an important role downstream of Cdc42 and Rac1 in cytoskeletal reorganization.
- Published
- 1998
47. PAK Kinases Are Directly Coupled to the PIX Family of Nucleotide Exchange Factors
- Author
-
Zhou-shen Zhao, Edward Manser, Thomas Leung, Lydia Tan, Ivan Tan, Cheng-Gee Koh, Louis Lim, Tsui-Han Loo, and Xiang-Qun Chen
- Subjects
Male ,rho GTP-Binding Proteins ,Membrane ruffling ,Molecular Sequence Data ,RAC1 ,Cell Cycle Proteins ,macromolecular substances ,GTPase ,Biology ,Protein Serine-Threonine Kinases ,Transfection ,environment and public health ,Second Messenger Systems ,SH3 domain ,Antibodies ,Gene Expression Regulation, Enzymologic ,src Homology Domains ,Mice ,PAK1 ,GTP-Binding Proteins ,Testis ,Cell Adhesion ,Animals ,Humans ,RNA, Messenger ,p21-activated kinases ,cdc42 GTP-Binding Protein ,Molecular Biology ,Sequence Homology, Amino Acid ,Kinase ,3T3 Cells ,Cell Biology ,Phosphoproteins ,Guanine Nucleotides ,Protein Structure, Tertiary ,Rats ,rac GTP-Binding Proteins ,Cytoskeletal Proteins ,Biochemistry ,p21-Activated Kinases ,Guanosine 5'-O-(3-Thiotriphosphate) ,Mutagenesis ,COS Cells ,Guanine nucleotide exchange factor ,Rabbits ,biological phenomena, cell phenomena, and immunity ,Paxillin ,HeLa Cells ,Protein Binding - Abstract
The PAK family of kinases are regulated through interaction with the small GTPases Cdc42 and Rac1, but little is known of the signaling components immediately upstream or downstream of these proteins. We have purified and cloned a new class of Rho-p21 guanine nucleotide exchange factor binding tightly through its N-terminal SH3 domain to a conserved proline-rich PAK sequence with a Kd of 24 nM. This PAK-interacting exchange factor (PIX), which is widely expressed and enriched in Cdc42- and Rac1-driven focal complexes, is required for PAK recruitment to these sites. PIX can induce membrane ruffling, with an associated activation of Rac1. Our results suggest a role for PIX in Cdc42-to-Rac1 signaling, involving the PIX/PAK complex.
- Published
- 1998
- Full Text
- View/download PDF
48. A Drosophila Homolog of the Rac- and Cdc42-Activated Serine/Threonine Kinase PAK Is a Potential Focal Adhesion and Focal Complex Protein That Colocalizes with Dynamic Actin Structures
- Author
-
Nicholas Harden, Yong-Mei Ong, Hui-Yong Loh, Ivan Tan, Thomas Leung, J. Lee, Edward Manser, and Louis Lim
- Subjects
Integrins ,Embryo, Nonmammalian ,Recombinant Fusion Proteins ,Molecular Sequence Data ,CDC42 ,Protein Serine-Threonine Kinases ,Biology ,Polymerase Chain Reaction ,Focal adhesion ,GTP-Binding Proteins ,Consensus Sequence ,Animals ,Drosophila Proteins ,Amino Acid Sequence ,Cloning, Molecular ,Phosphotyrosine ,Cytoskeleton ,p21-activated kinases ,Molecular Biology ,In Situ Hybridization ,DNA Primers ,Serine/threonine-specific protein kinase ,Base Sequence ,Sequence Homology, Amino Acid ,Cell Biology ,Actins ,Dorsal closure ,rac GTP-Binding Proteins ,Cell biology ,Rac GTP-Binding Proteins ,Drosophila melanogaster ,p21-Activated Kinases ,Epidermis ,Drosophila Protein ,Research Article ,Protein Binding - Abstract
Changes in cell morphology are essential in the development of a multicellular organism. The regulation of the cytoskeleton by the Rho subfamily of small GTP-binding proteins is an important determinant of cell shape. The Rho subfamily has been shown to participate in a variety of morphogenetic processes during Drosophila melanogaster development. We describe here a Drosophila homolog, DPAK, of the serine/threonine kinase PAK, a protein which is a target of the Rho subfamily proteins Rac and Cdc42. Rac, Cdc42, and PAK have previously been implicated in signaling by c-Jun amino-terminal kinases. DPAK bound to activated (GTP-bound) Drosophila Rac (DRacA) and Drosophila Cdc42. Similarities in the distributions of DPAK, integrin, and phosphotyrosine suggested an association of DPAK with focal adhesions and Cdc42- and Rac-induced focal adhesion-like focal complexes. DPAK was elevated in the leading edge of epidermal cells, whose morphological changes drive dorsal closure of the embryo. We have previously shown that the accumulation of cytoskeletal elements initiating cell shape changes in these cells could be inhibited by expression of a dominant-negative DRacA transgene. We show that leading-edge epidermal cells flanking segment borders, which express particularly large amounts of DPAK, undergo transient losses of cytoskeletal structures during dorsal closure. We propose that DPAK may be regulating the cytoskeleton through its association with focal adhesions and focal complexes and may be participating with DRacA in a c-Jun amino-terminal kinase signaling pathway recently demonstrated to be required for dorsal closure.
- Published
- 1996
49. Molecular Cloning of a New Member of the p21-Cdc42/Rac-activated Kinase (PAK) Family
- Author
-
Gregory J. Michael, Edward Manser, Zhuo-shen Zhao, Louis Lim, Christine Hall, Thomas Leung, and Claire Chong
- Subjects
rho GTP-Binding Proteins ,DNA, Complementary ,Molecular Sequence Data ,Cell Cycle Proteins ,macromolecular substances ,Protein Serine-Threonine Kinases ,Biology ,Mitogen-activated protein kinase kinase ,Biochemistry ,MAP2K7 ,GTP-Binding Proteins ,Animals ,Amino Acid Sequence ,RNA, Messenger ,c-Raf ,Cloning, Molecular ,Kinase activity ,cdc42 GTP-Binding Protein ,Molecular Biology ,Serine/threonine-specific protein kinase ,Base Sequence ,MAP kinase kinase kinase ,Cyclin-dependent kinase 4 ,Brain ,Cell Biology ,Molecular biology ,Recombinant Proteins ,Rats ,rac GTP-Binding Proteins ,p21-Activated Kinases ,biology.protein ,Cyclin-dependent kinase 9 ,Guanosine Triphosphate ,biological phenomena, cell phenomena, and immunity - Abstract
A number of "target" proteins for the Rho family of small GTP-binding proteins have now been identified, including the protein kinases ACK and p65PAK (Manser, E., Leung, T., Salihuddin, H., Zhao, Z.-S., and Lim, L. (1994) Nature 367, 40-46). The purified serine/threonine kinase p65PAK has been shown to be directly activated by GTP-Rac1 or GTP-Cdc42. Here we report the cDNA sequence encoding a new brain-enriched PAK isoform beta-PAK, which shares 79% amino acid identity with the previously described alpha-isoform. Their mRNAs are differentially expressed in the brain, with alpha-PAK mRNA being particularly abundant in motor-associated regions. In vitro translation products of the alpha- and beta-PAK cDNAs exhibited relative molecular masses of 68,000 and 65,000, respectively, by SDS-polyacrylamide analysis. A specific beta-PAK peptide sequence was obtained from rat brain-purified p65PAK. Recombinant alpha- and beta-PAKs exhibited an increase in kinase activity mediated by GTP-p21 induced autophosphorylation. Cdc42 was a more potent activator in vitro of alpha-PAK kinase, and the fully activated enzyme is 300 times more active than the unphosphorylated form. Interestingly the down-regulation in the binding of p21s to recombinant beta-PAK and brain p65PAK, which is observed upon kinase activation does not occur with recombinant alpha-PAK.
- Published
- 1995
50. The GTPase-deficient Rnd proteins are stabilized by their effectors
- Author
-
Liuh Ling Goh and Edward Manser
- Subjects
rac1 GTP-Binding Protein ,rho GTP-Binding Proteins ,RHOA ,Leupeptins ,CDC42 ,GTPase ,Biology ,Protein degradation ,Cysteine Proteinase Inhibitors ,Biochemistry ,Chlorocebus aethiops ,Enzyme Stability ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,cdc42 GTP-Binding Protein ,Molecular Biology ,Zebrafish ,rho-Associated Kinases ,Rnd3 ,Effector ,Cell Biology ,Protein-Tyrosine Kinases ,Cell biology ,Protein Structure, Tertiary ,Repressor Proteins ,HEK293 Cells ,Cdc42 GTP-Binding Protein ,p21-Activated Kinases ,Protein Synthesis and Degradation ,COS Cells ,Mutation ,biology.protein ,Guanine nucleotide exchange factor ,HeLa Cells - Abstract
Rnd proteins are Rho family GTP-binding proteins with cellular functions that antagonize RhoA signaling. We recently described a new Rnd3 effector Syx, also named PLEKHG5, that interacts with Rnds via a Raf1-like "Ras-binding domain." Syx is a multidomain RhoGEF that participates in early zebrafish development. Here we demonstrated that Rnd1, Rnd2, and Rnd3 stability is acutely dependent on interaction with their effectors such as Syx or p190 RhoGAP. Although Rnd3 turnover is blocked by treatment of cells with MG132, we provide evidence that such turnover is mediated indirectly by effects on the Rnd3 effectors, rather than on Rnd3 itself, which is not significantly ubiquitinated. The minimal regions of Syx and p190 RhoGAP that bind Rnd3 are not sequence-related but have similar effects. We have identified features that allow for Rnd3 turnover including a conserved Lys-45 close to the switch I region and the C-terminal membrane-binding domain of Rnd3, which cannot be substituted by the equivalent Cdc42 CAAX sequence. By contrast, an effector binding-defective mutant of Rnd3 when overexpressed undergoes turnover at normal rates. Interestingly the activity of the RhoA-regulated kinase ROCK stimulates Rnd3 turnover. This study suggests that Rnd proteins are regulated through feedback mechanisms in cells where the level of effectors and RhoA activity influence the stability of Rnd proteins. This effector feedback behavior is analogous to the ability of ACK1 and PAK1 to prolong the lifetime of the active GTP-bound state of Cdc42 and Rac1.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.