Network


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

Hotspot


Dive into the research topics where Grant Scotland is active.

Publication


Featured researches published by Grant Scotland.


Journal of Biological Chemistry | 1999

The RACK1 Signaling Scaffold Protein Selectively Interacts with the cAMP-specific Phosphodiesterase PDE4D5 Isoform

Stephen J. Yarwood; Michael R. Steele; Grant Scotland; Miles D. Houslay; Graeme B. Bolger

The WD-repeat protein receptor for activated C-kinase (RACK1) was identified by its interaction with the cyclic AMP-specific phosphodiesterase (PDE4) isoform PDE4D5 in a yeast two-hybrid screen. The interaction was confirmed by co-immunoprecipitation of native RACK1 and PDE4D5 from COS7, HEK293, 3T3-F442A, and SK-N-SH cell lines. The interaction was unaffected by stimulation of the cells with the phorbol ester phorbol 2-myristate 3-acetate. PDE4D5 did not interact with two other WD-repeat proteins, β’-coatomer protein and Gsβ, in two-hybrid tests. RACK1 did not interact with other PDE4D isoforms or with known PDE4A, PDE4B, and PDE4C isoforms. PDE4D5 and RACK1 interacted with high affinity (K a approximately 7 pm) when they were expressed and purified from Escherichia coli, demonstrating that the interaction does not require intermediate proteins. The binding of the E. coli-expressed proteins did not alter the kinetics of cAMP hydrolysis by PDE4D5 but caused a 3–4-fold change in its sensitivity to inhibition by the PDE4 selective inhibitor rolipram. The subcellular distributions of RACK1 and PDE4D5 were extremely similar, with the major amount of both proteins (70%) in the high speed supernatant (S2) fraction. Analysis of constructs with specific deletions or single amino acid mutations in PDE4D5 demonstrated that a small cluster of amino acids in the unique amino-terminal region of PDE4D5 was necessary for its interaction with RACK1. We suggest that RACK1 may act as a scaffold protein to recruit PDE4D5 and other proteins into a signaling complex.


Journal of Biological Chemistry | 1999

Association with the SRC Family Tyrosyl Kinase LYN Triggers a Conformational Change in the Catalytic Region of Human cAMP-specific Phosphodiesterase HSPDE4A4B CONSEQUENCES FOR ROLIPRAM INHIBITION

Ian McPhee; Stephen J. Yarwood; Grant Scotland; Elaine Huston; M. Beard; Annette H. Ross; Emma S. Houslay; Miles D. Houslay

The cAMP-specific phosphodiesterase (PDE) HSPDE 4A4B(pde46) selectively bound SH3 domains of SRC family tyrosyl kinases. Such an interaction profoundly changed the inhibition of PDE4 activity caused by the PDE4-selective inhibitor rolipram and mimicked the enhanced rolipram inhibition seen for particulate, compared with cytosolic pde46 expressed in COS7 cells. Particulate pde46 co-localized with LYN kinase in COS7 cells. The unique N-terminal and LR2 regions of pde46 contained the sites for SH3 binding. Altered rolipram inhibition was triggered by SH3 domain interaction with the LR2 region. Purified LYN SH3 and human PDE4A LR2 could be co-immunoprecipitated, indicating a direct interaction. Protein kinase A-phosphorylated pde46 remained able to bind LYN SH3. pde46 was found to be associated with SRC kinase in the cytosol of COS1 cells, leading to aberrant kinetics of rolipram inhibition. It is suggested that pde46 may be associated with SRC family tyrosyl kinases in intact cells and that the ensuing SH3 domain interaction with the LR2 region of pde46 alters the conformation of the PDE catalytic unit, as detected by altered rolipram inhibition. Interaction between pde46 and SRC family tyrosyl kinases highlights a potentially novel regulatory system and point of signaling system cross-talk.


Journal of Biological Chemistry | 1996

The Human Cyclic AMP-specific Phosphodiesterase PDE-46 (HSPDE4A4B) Expressed in Transfected COS7 Cells Occurs as Both Particulate and Cytosolic Species That Exhibit Distinct Kinetics of Inhibition by the Antidepressant Rolipram

Elaine Huston; Linda Pooley; Pascale Julien; Grant Scotland; Ian McPhee; Michael Sullivan; Graeme B. Bolger; Miles D. Houslay

Transfection of COS7 cells with a plasmid encoding the human cyclic AMP-specific PDE4A phosphodiesterase PDE-46 (HSPDE4A4B) led to the expression of a rolipram-inhibited PDE4 activity, which contributed ∼96% of the total COS cell PDE activity. A fusion protein was generated which encompassed residues (788-886) at the extreme C terminus of PDE-46 and was used to generate an antiserum that detected PDE-46 in transfected COS7 cells. Immunoblotting studies identified PDE-46 as a ∼125-kDa species that was associated with both the soluble and particulate fractions. The relative Vmax of particulate PDE-46 was ∼56% that of cytosolic PDE-46. Particulate PDE-46 was not solubilized using Triton X-100 or high NaCl concentrations. Immunofluorescence analysis by laser scanning confocal microscopy showed that PDE-46 was located at discrete margins of the cell, indicative of association with membrane cortical regions. The human PDE4A species, h6.1 (HSPDE4A4C), which lacks the N-terminal extension of PDE-46, was found as an entirely soluble species when expressed in COS7 cells. h6.1 was shown to have an ∼11-fold higher Vmax relative to that of PDE-46. In dose-response studies rolipram inhibited particulate PDE-46 at much lower concentrations (IC50 = 0.195 μM) than those needed to inhibit the cytosolic enzyme (IC50 = 1.6 μM). The basis of this difference lay in the fact that rolipram served as a simple competitive inhibitor of the cytosol enzyme (Ki = 1.6 μM) but as a partial competitive inhibitor of the particulate enzyme (Ki = 0.037 μM; Ki′ = 2.3 μM). Particulate PDE-46 thus showed a ∼60-fold higher affinity for rolipram than cytosolic PDE-46.


Journal of Biological Chemistry | 1996

Determination of the Structure of the N-terminal Splice Region of the Cyclic AMP-specific Phosphodiesterase RD1 (RNPDE4A1) by 1H NMR and Identification of the Membrane Association Domain Using Chimeric Constructs

Kate Smith; Grant Scotland; J Beattie; Ian P. Trayer; Houslay

A 25-residue peptide representing the membrane targeting N-terminal splice region of the cyclic AMP phosphodiesterase RD1 (RNPDE4A1) was synthesized, and its structure was determined by 1H NMR. Two independently folding helical regions were identified, separated by a highly mobile “hinge” region. The first helical region was formed by an N-terminal amphipathic α-helix, and the second consisted of multiple overlapping turns and contained a distinct compact, hydrophobic, tryptophan-rich domain (residues 14-20). Chimeric molecules, formed between the N-terminal region of RD1 and the soluble bacterial protein chloramphenicol acetyltransferase, were used in an in vitro system to determine the features within the splice region that were required for membrane association. The ability of RD1-chloramphenicol acetyltransferase chimera to become membrane-associated was not affected by deletion of any of the following regions: the apolar section (residues 2-7) of the first helical region, the polar part of this region together with the hinge region (residues 8-13), or the polar end of the C-terminal helical region (residues 21-25). In marked contrast, deletion of the compact, hydrophobic tryptophan-rich domain (residues 14-20) found in the second helical region obliterated membrane association. Replacement of this domain with a hydrophobic cassette of seven alanine residues also abolished membrane association, indicating that membrane-association occurred by virtue of specific hydrophobic interactions with residues within the compact, tryptophan-rich domain. The structure of this domain is well defined in the peptide, and although the region is helical, both the backbone and the distribution of side chains are somewhat distorted as compared with an ideal α-helix. Hydrophobic interactions, such as the “stacked” rings of residues Pro14 and Trp15, stabilize this domain with the side chain of residue Leu16 adopting a central position, interacting with the side chains of all three tryptophan residues 15, 19, and 20. These bulky side chains thus form a hydrophobic cluster. In contrast, the side chain of residue Val17 is relatively exposed, pointing out from the opposite “face” of the peptide. Although it appears that this compact, tryptophan-rich domain is responsible for membrane association, at present the target site and hence the specific interactions involved in membrane targeting by the RD1 splice region remain unidentified.


Biochemical Journal | 1997

Characterization of five different proteins produced by alternatively spliced mRNAs from the human cAMP-specific phosphodiesterase PDE4D gene.

Graeme B. Bolger; Suat Erdogan; Randy E. Jones; Kate Loughney; Grant Scotland; Ralf Hoffmann; Ian Wilkinson; Cathy Farrell; Miles D. Houslay


Biochemical Journal | 1995

Chimeric constructs show that the unique N-terminal domain of the cyclic AMP phosphodiesterase RD1 (RNPDE4A1A; rPDE-IVA1) can confer membrane association upon the normally cytosolic protein chloramphenicol acetyltransferase.

Grant Scotland; Miles D. Houslay


Journal of Biological Chemistry | 2000

The cAMP-specific Phosphodiesterase PDE4A5 Is Cleaved Downstream of Its SH3 Interaction Domain by Caspase-3 CONSEQUENCES FOR ALTERED INTRACELLULAR DISTRIBUTION

Elaine Huston; M. Beard; Fraser McCallum; Nigel J. Pyne; Peter Vandenabeele; Grant Scotland; Miles D. Houslay


Cellular Signalling | 2002

In addition to the SH3 binding region, multiple regions within the N-terminal noncatalytic portion of the cAMP-specific phosphodiesterase, PDE4A5, contribute to its intracellular targeting

M. Beard; Elaine Huston; Lachlan Campbell; Irene Gall; Ian McPhee; Stephen J. Yarwood; Grant Scotland; Miles D. Houslay


Biochemical Society Transactions | 1997

Intracellular targeting, interaction with Src homology 3 (SH3) domains and rolipram-detected conformational switches in cAMP-specific PDE4A phosphodiesterase

Miles D. Houslay; Grant Scotland; Suat Erdogan; Elaine Huston; Simon Mackenzie; J F McCallum; Ian McPhee; Linda Pooley; Graham Rena; Annette H. Ross; M. Beard; A. Peder; F. Begg; I. Wilkinson; Stephen J. Yarwood; C. Ackerman; Emma S. Houslay; R. Hoffman; Paul T. Engels; Michael Sullivan; Graeme B. Bolger


Methods | 1998

Intracellular Compartmentalization of PDE4 Cyclic AMP-Specific Phosphodiesterases☆

Grant Scotland; M. Beard; Suat Erdogan; Elaine Huston; F. McCallum; Simon J. MacKenzie; A.H. Peden; Linda Pooley; N.G. Rena; Annette H. Ross; Stephen J. Yarwood; Miles D. Houslay

Collaboration


Dive into the Grant Scotland's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Beard

University of Glasgow

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Graeme B. Bolger

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge