Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Edward Manser is active.

Publication


Featured researches published by Edward Manser.


Molecular Cell | 1998

PAK Kinases Are Directly Coupled to the PIX Family of Nucleotide Exchange Factors

Edward Manser; Tsui-Han Loo; Cheng-Gee Koh; Zhou-shen Zhao; Xiang-Qun Chen; Lydia Tan; Ivan Tan; Thomas Leung; Louis Lim

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.


Molecular and Cellular Biology | 1998

A Conserved Negative Regulatory Region in αPAK: Inhibition of PAK Kinases Reveals Their Morphological Roles Downstream of Cdc42 and Rac1

Zhou-shen Zhao; Edward Manser; Xiang-Qun Chen; Claire Chong; Thomas Leung; Louis Lim

ABSTRACT αPAK 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 αPAK). αPAK 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 αPAK83-149 potently inhibited GTPγS-bound Cdc42-mediated kinase activation of both αPAK and βPAK. 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 αPAK83-149 completely blocked the phenotypic effects of hyperactive αPAKL107F 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.


Biochemical Journal | 2005

PAK and other Rho-associated kinases--effectors with surprisingly diverse mechanisms of regulation.

Zhou-shen Zhao; Edward Manser

The Rho GTPases are a family of molecular switches that are critical regulators of signal transduction pathways in eukaryotic cells. They are known principally for their role in regulating the cytoskeleton, and do so by recruiting a variety of downstream effector proteins. Kinases form an important class of Rho effector, and part of the biological complexity brought about by switching on a single GTPase results from downstream phosphorylation cascades. Here we focus on our current understanding of the way in which different Rho-associated serine/threonine kinases, denoted PAK (p21-activated kinase), MLK (mixed-lineage kinase), ROK (Rho-kinase), MRCK (myotonin-related Cdc42-binding kinase), CRIK (citron kinase) and PKN (protein kinase novel), interact with and are regulated by their partner GTPases. All of these kinases have in common an ability to dimerize, and in most cases interact with a variety of other proteins that are important for their function. A diversity of known structures underpin the Rho GTPase-kinase interaction, but only in the case of PAK do we have a good molecular understanding of kinase regulation. The ability of Rho GTPases to co-ordinate spatial and temporal phosphorylation events explains in part their prominent role in eukaryotic cell biology.


The EMBO Journal | 1998

PAK promotes morphological changes by acting upstream of Rac

Axel Obermeier; Sohail Ahmed; Edward Manser; Su Ching Yen; Christine Hall; Louis Lim

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 βPAK 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.


Journal of Biological Chemistry | 2001

The Tyrosine Kinase ACK1 Associates with Clathrin-coated Vesicles through a Binding Motif Shared by Arrestin and Other Adaptors

Mabel Teo; Lydia Tan; Louis Lim; Edward Manser

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 β-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 ACK1in vivo with clathrin and AP-2 indicates that it participates in trafficking, underlying an ability to increase receptor-mediated transferrin uptake.


Genes to Cells | 2002

Phosphorylation and reorganization of vimentin by p21-activated kinase (PAK)

Hidemasa Goto; Kazushi Tanabe; Edward Manser; Louis Lim; Yoshihiro Yasui; Masaki Inagaki

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.


Progress in molecular and subcellular biology | 1999

Roles of PAK Family Kinases

Edward Manser; Louis Lim

The first identified protein kinase exhibiting direct activation by a Ras-related GTPase was a brain p21(Cdc42/Rac) activated kinase designated p65PAK(Manser et al 1994).The kinase(s) were originally identified in [γ32P]GTPCdc42 and [γ32P]GTP-Rac1 overlays of SDS-polyacrylamide gel fractionated brain proteins(Manser et al. 1993). The presence of similar sized target proteins in all tissues suggested that these serine/threonine protein kinases were ubiquitous. In binding the active GTP-forms of Cdc42 and Racl, the α-and γ-PAKs also decrease intrinsic GTP hydrolysis and block action of GTPase activating proteins (GAPs). This therefore favours signalling to PAK. However, βPAK may be released after activation allowing for amplification of the signal or initiation of other p21 actions(Manser et al 1995, Manser et al 1994).


Biochemical Journal | 2009

Paxillin nuclear-cytoplasmic localization is regulated by phosphorylation of the LD4 motif: evidence that nuclear paxillin promotes cell proliferation

Jing-Ming Dong; Lei-Shong Lau; Yuen-Wai Ng; Louis Lim; Edward Manser

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 LD(4) motif that binds GIT1 and FAK1 (focal-adhesion kinase 1). We provide evidence that phosphorylation of Ser(272) within LD(4) blocks nuclear export, and we show that this modification also reduces GIT1, but not FAK1, binding; however, Ser(272) 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.


FEBS Letters | 2005

SNX9 as an adaptor for linking synaptojanin‐1 to the Cdc42 effector ACK1

Lee Yeow-Fong; Louis Lim; Edward Manser

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.


European Journal of Cell Biology | 2002

Vimentin intermediate filament reorganization by Cdc42: involvement of PAK and p70 S6 kinase.

Wing Chan; Robert Kozma; Yoshihiro Yasui; Masaki Inagaki; Thomas Leung; Edward Manser; Louis Lim

Rho family GTPases play a major role in actin cytoskeleton reorganization. Recent studies have shown that the activation of Rho family GTPases also induces collapse of the vimentin intermediate filament (IF) network in fibroblasts. Here, we report that Cdc42V12 induces the reorganization of vimentin IFs in Hela cells, and such reorganization is independent of actin and microtubule status. We analyzed the involvement of three serine/threonine kinase effectors, MRCK, PAK and p70 S6K in the Cdc42-induced vimentin reorganization. Surprisingly, the ROK-related MRCK is not involved in this IF reorganization. We detected phosphorylation of vimentin Ser72, a site phosphorylated by PAK, after Cdc42 activation. PAK inhibition partially blocked Cdc42-induced vimentin IF collapse suggesting the involvement of other effectors. We report that p70 S6 kinase (S6K)1 participates in this IF rearrangement since the inhibitor rapamycin or a dominant inhibitory S6K could reduce the Cdc42V12 or bradykinin-induced vimentin collapse. Further, inhibition of PAK and S6K in combination very effectively prevents Cdc42-induced vimentin IF collapse. Conversely, only in combination active PAK and S6K could induce a vimentin IF rearrangement that mimics the Cdc42 effect. Thus, Cdc42-induced vimentin reorganization involves PAK and, in a novel cytoskeletal role, p70 S6K.

Collaboration


Dive into the Edward Manser's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Leung

Institute of Molecular and Cell Biology

View shared research outputs
Top Co-Authors

Avatar

Zhou-shen Zhao

Institute of Molecular and Cell Biology

View shared research outputs
Top Co-Authors

Avatar

Lydia Tan

Institute of Molecular and Cell Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cheng-Gee Koh

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar

Claire Chong

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Jing-Ming Dong

Institute of Molecular and Cell Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge