Network


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

Hotspot


Dive into the research topics where Heidi C. E. Welch is active.

Publication


Featured researches published by Heidi C. E. Welch.


FEBS Letters | 2003

Phosphoinositide 3‐kinase‐dependent activation of Rac

Heidi C. E. Welch; W. John Coadwell; Len Stephens; Phillip T. Hawkins

The monomeric GTPase Rac and the lipid kinase phosphoinositide 3‐kinase (PI3K) are intracellular signalling enzymes that each regulate a huge range of cellular functions. Their signalling pathways overlap. Several pathways lead from PI3K activation via the production of the lipid second messenger phosphatidylinositol (3,4,5)‐triphosphate (PtdIns(3,4,5)P3) to the activation of guanine‐nucleotide exchange factors (GEFs) that activate Rac. Vice versa, Rac can also stimulate the activation of PI3K, although the mechanism for this is unclear. We review here the evidence that links PI3K and Rac signalling pathways.


Current Biology | 2005

P-Rex1 Regulates Neutrophil Function

Heidi C. E. Welch; Alison M. Condliffe; Laura Milne; G. John Ferguson; Kirsti Hill; Louise M. C. Webb; Klaus Okkenhaug; W. John Coadwell; Simon Andrews; Marcus Thelen; Gareth E. Jones; Phillip T. Hawkins; Len Stephens

Rac GTPases regulate cytoskeletal structure, gene expression, and reactive oxygen species (ROS) production. Rac2-deficient neutrophils cannot chemotax, produce ROS, or degranulate upon G protein-coupled receptor (GPCR) activation. Deficiency in PI3Kgamma, an upstream regulator of Rac, causes a similar phenotype. P-Rex1, a guanine-nucleotide exchange factor (GEF) for Rac, is believed to link GPCRs and PI3Kgamma to Rac-dependent neutrophil responses. We have investigated the functional importance of P-Rex1 by generating a P-Rex1(-/-) mouse. P-Rex1(-/-) mice are viable and healthy, with apparently normal leukocyte development, but with mild neutrophilia. In neutrophils from P-Rex1(-/-) mice, GPCR-dependent Rac2 activation is impaired, whereas Rac1 activation is less compromised. GPCR-dependent ROS formation is absent in lipopolysaccharide (LPS)-primed P-Rex1(-/-) neutrophils, but less affected in unprimed or TNFalpha-primed cells. Recruitment of P-Rex1(-/-) neutrophils to inflammatory sites is impaired. Surprisingly, chemotaxis of isolated neutrophils is only slightly reduced, with a mild defect in cell speed, but normal polarization and directionality. Secretion of azurophil granules is unaffected. In conclusion, P-Rex1 is an important regulator of neutrophil function by mediating a subset of Rac-dependent neutrophil responses. However, P-Rex1 is not an essential regulator of neutrophil chemotaxis and degranulation.


Nature Cell Biology | 2006

Gβγs and the Ras binding domain of p110γ are both important regulators of PI3Kγ signalling in neutrophils

Sabine Suire; Alison M. Condliffe; G. John Ferguson; Chris D. Ellson; Hervé Guillou; Keith Davidson; Heidi C. E. Welch; John Coadwell; Martin Turner; Edwin R. Chilvers; Phillip T. Hawkins; Len Stephens

Through their ability to regulate production of the key lipid messenger PtdIns(3,4,5)P3, the class I phosphatidylinositol-3-OH kinases (PI(3)Ks) support many critical cell responses. They, in turn, can be regulated by cell-surface receptors through signals acting on either their adaptor subunits (for example, through phosphotyrosine or Gβγs) or their catalytic subunits (for example, through GTP-Ras). The relative significance of these controlling inputs is undefined in vivo. Here, we have studied the roles of Gβγs, the adaptor p101, Ras and the Ras binding domain (RBD) in the control of the class I PI(3)K, PI(3)Kγ, in mouse neutrophils. Loss of p101 leads to major reductions in the accumulation of PtdIns(3,4,5)P3, activation of protein kinase B (PKB) and in migration towards G-protein activating ligands in vitro, and to an aseptically inflamed peritoneum in vivo. Loss of sensitivity of PI(3)Kγ to Ras unexpectedly caused similar reductions, but additionally caused a substantial loss in production of reactive oxygen species (ROS). We conclude that Gβγs, p101 and the Ras–RBD interaction all have important roles in the regulation of PI(3)Kγ in vivo and that they can simultaneously, but differentially, control distinct PI(3)Kγ effectors.


Nature Communications | 2011

P-Rex1 is required for efficient melanoblast migration and melanoma metastasis

Colin R. Lindsay; Samuel Lawn; Andrew D. Campbell; William J. Faller; Florian Rambow; Richard L. Mort; Paul Timpson; Ang Li; Patrizia Cammareri; Rachel A. Ridgway; Jennifer P. Morton; Brendan Doyle; Shauna Hegarty; Mairin Rafferty; Ian Murphy; Enda W. McDermott; Kieran Sheahan; Katherine H. Pedone; Alexander J. Finn; Pamela A. Groben; Nancy E. Thomas; Honglin Hao; Craig Carson; Jim C. Norman; Laura M. Machesky; William M. Gallagher; Ian J. Jackson; Leon Van Kempen; Friedrich Beermann; Channing J. Der

Metastases are the major cause of death from melanoma, a skin cancer that has the fastest rising incidence of any malignancy in the Western world. Molecular pathways that drive melanoblast migration in development are believed to underpin the movement and ultimately the metastasis of melanoma. Here we show that mice lacking P-Rex1, a Rac-specific Rho GTPase guanine nucleotide exchange factor, have a melanoblast migration defect during development evidenced by a white belly. Moreover, these P-Rex1(-/-) mice are resistant to metastasis when crossed to a murine model of melanoma. Mechanistically, this is associated with P-Rex1 driving invasion in a Rac-dependent manner. P-Rex1 is elevated in the majority of human melanoma cell lines and tumour tissue. We conclude that P-Rex1 has an important role in melanoblast migration and cancer progression to metastasis in mice and humans.


Journal of Biological Chemistry | 1998

PROTEIN KINASE B AND RAC ARE ACTIVATED IN PARALLEL WITHIN A PHOSPHATIDYLINOSITIDE 3OH-KINASE-CONTROLLED SIGNALING PATHWAY

Heidi C. E. Welch; Alicia Eguinoa; Leonard R. Stephens; Phillip T. Hawkins

The GTPase Rac and the protein kinase B (PKB) are downstream targets of phosphatidylinositide 3OH-kinase in platelet-derived growth factor-stimulated signaling pathways. We have generated PAE cell lines inducibly expressing mutants of Rac. Use of these cell lines suggests that Rac is involved in both platelet-derived growth factor-stimulated membrane ruffling and the activation of p70S6K but not in the activation of PKB. Furthermore, expression of constitutively active alleles of PKB in PAE cells suggests that PKB is able to regulate the activity of p70S6K but not the cytoskeletal changes underlying membrane ruffling. Thus, our results indicate that Rac and PKB are on separate pathways downstream of phosphatidylinositide 3OH-kinase in these cells but that both of these pathways are involved in the regulation of p70S6K.


FEBS Letters | 2004

P-Rex2, a new guanine-nucleotide exchange factor for Rac

Sarah Donald; Kirsti Hill; Charlotte Lécureuil; Romain Barnouin; Sonja Krugmann; W. John Coadwell; Simon Andrews; Simon Walker; Phillip T. Hawkins; Len Stephens; Heidi C. E. Welch

We have identified a new guanine‐nucleotide exchange factor, P‐Rex2, and cloned it from human skeletal muscle and brain libraries. It has widespread tissue distribution but is not expressed in neutrophils. P‐Rex2 is a 183 kDa protein that activates the small GTPase Rac and is regulated by phosphatidylinositol (3,4,5)‐trisphosphate and the βγ subunits of heterotrimeric G proteins in vitro and in vivo. P‐Rex2 has structure, activity and regulatory properties similar to P‐Rex1 but has divergent tissue distribution, as P‐Rex1 is mainly expressed in neutrophils. Together, they form an enzyme family capable of mediating Rac signalling downstream of G protein‐coupled receptors and phosphoinositide 3‐kinase.


Journal of Biological Chemistry | 2007

Membrane Translocation of P-Rex1 Is Mediated by G Protein βγ Subunits and Phosphoinositide 3-Kinase

Mark A. Barber; Sarah Donald; Sylvia Thelen; Karen E. Anderson; Marcus Thelen; Heidi C. E. Welch

P-Rex1 is a guanine-nucleotide exchange factor (GEF) for the small GTPase Rac that is directly activated by the βγ subunits of heterotrimeric G proteins and by the lipid second messenger phosphatidylinositol (3,4,5)-trisphosphate (PIP3), which is generated by phosphoinositide 3-kinase (PI3K). Gβγ subunits and PIP3 are membrane-bound, whereas the intracellular localization of P-Rex1 in basal cells is cytosolic. Activation of PI3K alone is not sufficient to promote significant membrane translocation of P-Rex1. Here we investigated the subcellular localization of P-Rex1 by fractionation of Sf9 cells co-expressing P-Rex1 with Gβγ and/or PI3K. In basal, serum-starved cells, P-Rex1 was mainly cytosolic, but 7% of the total was present in the 117,000 × g membrane fraction. Co-expression of P-Rex1 with either Gβγ or PI3K caused only an insignificant increase in P-Rex1 membrane localization, whereas Gβγ and PI3K together synergistically caused a robust increase in membrane-localized P-Rex1 to 23% of the total. PI3K-driven P-Rex1 membrane recruitment was wortmannin-sensitive. The use of P-Rex1 mutants showed that the isolated Dbl homology/pleckstrin homology domain tandem of P-Rex1 is sufficient for synergistic Gβγ- and PI3K-driven membrane localization; that the enzymatic GEF activity of P-Rex1 is not required for membrane translocation; and that the other domains of P-Rex1 (DEP, PDZ, and IP4P) contribute to keeping the enzyme localized in the cytosol of basal cells. In vitro Rac2-GEF activity assays showed that membrane-derived purified P-Rex1 has a higher basal activity than cytosol-derived P-Rex1, but both can be further activated by PIP3 and Gβγ subunits.


Proceedings of the National Academy of Sciences of the United States of America | 2008

P-Rex2 regulates Purkinje cell dendrite morphology and motor coordination

Sarah Donald; Trevor Humby; Ian Fyfe; Anne Segonds-Pichon; Simon Walker; Simon Andrews; W. John Coadwell; Piers C. Emson; Lawrence Stephen Wilkinson; Heidi C. E. Welch

The small GTPase Rac controls cell morphology, gene expression, and reactive oxygen species formation. Manipulations of Rac activity levels in the cerebellum result in motor coordination defects, but activators of Rac in the cerebellum are unknown. P-Rex family guanine-nucleotide exchange factors activate Rac. We show here that, whereas P-Rex1 expression within the brain is widespread, P-Rex2 is specifically expressed in the Purkinje neurons of the cerebellum. We have generated P-Rex2−/− and P-Rex1−/−/P-Rex2−/− mice, analyzed their Purkinje cell morphology, and assessed their motor functions in behavior tests. The main dendrite is thinned in Purkinje cells of P-Rex2−/− pups and dendrite structure appears disordered in Purkinje cells of adult P-Rex2−/− and P-Rex1−/−/P-Rex2−/− mice. P-Rex2−/− mice show a mild motor coordination defect that progressively worsens with age and is more pronounced in females than in males. P-Rex1−/−/P-Rex2−/− mice are ataxic, with reduced basic motor activity and abnormal posture and gait, as well as impaired motor coordination even at a young age. We conclude that P-Rex1 and P-Rex2 are important regulators of Purkinje cell morphology and cerebellar function.


Journal of Immunology | 2011

P-Rex1 and Vav1 Cooperate in the Regulation of Formyl-Methionyl-Leucyl-Phenylalanine–Dependent Neutrophil Responses

Campbell D. Lawson; Sarah Donald; Karen E. Anderson; Daniel T. Patton; Heidi C. E. Welch

G protein-coupled receptor (GPCR) activation elicits neutrophil responses such as chemotaxis and reactive oxygen species (ROS) formation, which depend on the small G protein Rac and are essential for host defense. P-Rex and Vav are two families of guanine-nucleotide exchange factors (GEFs) for Rac, which are activated through distinct mechanisms but can both control GPCR-dependent neutrophil responses. It is currently unknown whether they play specific roles or whether they can compensate for each other in controlling these responses. In this study, we have assessed the function of neutrophils from mice deficient in P-Rex and/or Vav family GEFs. We found that both the P-Rex and the Vav family are important for LPS priming of ROS formation, whereas particle-induced ROS responses and cell spreading are controlled by the Vav family alone. Surprisingly, fMLF-stimulated ROS formation, adhesion, and chemotaxis were synergistically controlled by P-Rex1 and Vav1. These responses were more severely impaired in neutrophils lacking both P-Rex1 and Vav1 than those lacking the entire P-Rex family, the entire Vav family, or both P-Rex1 and Vav3. P-Rex1/Vav1 (P1V1) double-deficient cells also showed the strongest reduction in fMLF-stimulated activation of Rac1 and Rac2. This reduction in Rac activity may be sufficient to cause the defects observed in fMLF-stimulated P1V1 neutrophil responses. Additionally, Mac-1 surface expression was reduced in P1V1 cells, which might contribute further to defects in responses involving integrins, such as GPCR-stimulated adhesion and chemotaxis. We conclude that P-Rex1 and Vav1 together are the major fMLFR -dependent Dbl family Rac-GEFs in neutrophils and cooperate in the control of fMLF-stimulated neutrophil responses.


Cell Reports | 2014

The Rac-FRET Mouse Reveals Tight Spatiotemporal Control of Rac Activity in Primary Cells and Tissues

Anna-Karin E. Johnsson; Yanfeng Dai; Max Nobis; Martin J. Baker; Ewan J. McGhee; Simon Walker; Juliane P. Schwarz; Shereen Kadir; Jennifer P. Morton; Kevin Myant; David J. Huels; Anne Segonds-Pichon; Owen J. Sansom; Kurt I. Anderson; Paul Timpson; Heidi C. E. Welch

Summary The small G protein family Rac has numerous regulators that integrate extracellular signals into tight spatiotemporal maps of its activity to promote specific cell morphologies and responses. Here, we have generated a mouse strain, Rac-FRET, which ubiquitously expresses the Raichu-Rac biosensor. It enables FRET imaging and quantification of Rac activity in live tissues and primary cells without affecting cell properties and responses. We assessed Rac activity in chemotaxing Rac-FRET neutrophils and found enrichment in leading-edge protrusions and unexpected longitudinal shifts and oscillations during protruding and stalling phases of migration. We monitored Rac activity in normal or disease states of intestinal, liver, mammary, pancreatic, and skin tissue, in response to stimulation or inhibition and upon genetic manipulation of upstream regulators, revealing unexpected insights into Rac signaling during disease development. The Rac-FRET strain is a resource that promises to fundamentally advance our understanding of Rac-dependent responses in primary cells and native environments.

Collaboration


Dive into the Heidi C. E. Welch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul Timpson

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge