Network


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

Hotspot


Dive into the research topics where Alan J. Whitmarsh is active.

Publication


Featured researches published by Alan J. Whitmarsh.


The EMBO Journal | 1996

Selective interaction of JNK protein kinase isoforms with transcription factors

Shashi Gupta; Tamera Barrett; Alan J. Whitmarsh; Julie Cavanagh; Hayla Karen Sluss; Benoit Derijard; Roger J. Davis

The JNK protein kinase is a member of the MAP kinase group that is activated in response to dual phosphorylation on threonine and tyrosine. Ten JNK isoforms were identified in human brain by molecular cloning. These protein kinases correspond to alternatively spliced isoforms derived from the JNK1, JNK2 and JNK3 genes. The protein kinase activity of these JNK isoforms was measured using the transcription factors ATF2, Elk‐1 and members of the Jun family as substrates. Treatment of cells with interleukin‐1 (IL‐1) caused activation of the JNK isoforms. This activation was blocked by expression of the MAP kinase phosphatase MKP‐1. Comparison of the binding activity of the JNK isoforms demonstrated that the JNK proteins differ in their interaction with ATF2, Elk‐1 and Jun transcription factors. Individual members of the JNK group may therefore selectively target specific transcription factors in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2003

A critical role of neural-specific JNK3 for ischemic apoptosis.

Chia Yi Kuan; Alan J. Whitmarsh; Derek D. Yang; Guanghong Liao; Aryn Schloemer; Chen Dong; Jue Bao; Kenneth J Banasiak; Gabriel G. Haddad; Richard A. Flavell; Roger J. Davis; Pasko Rakic

c-Jun N-terminal kinase (JNK) signaling is an important contributor to stress-induced apoptosis, but it is unclear whether JNK and its isoforms (JNK1, JNK2, and JNK3) have distinct roles in cerebral ischemia. Here we show that JNK1 is the major isoform responsible for the high level of basal JNK activity in the brain. In contrast, targeted deletion of Jnk3 not only reduces the stress-induced JNK activity, but also protects mice from brain injury after cerebral ischemia–hypoxia. The downstream mechanism of JNK3-mediated apoptosis may include the induction of Bim and Fas and the mitochondrial release of cytochrome c. These results suggest that JNK3 is a potential target for neuroprotection therapies in stroke.


Nature Genetics | 2006

Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch

Rebecca J. Richardson; Jill Dixon; Saimon Malhotra; Matthew J. Hardman; Lynnette Knowles; Ray Boot-Handford; Paul Shore; Alan J. Whitmarsh; Michael J. Dixon

The epidermis is a highly organized structure, the integrity of which is central to the protection of an organism. Development and subsequent maintenance of this tissue depends critically on the intricate balance between proliferation and differentiation of a resident stem cell population; however, the signals controlling the proliferation-differentiation switch in vivo remain elusive. Here, we show that mice carrying a homozygous missense mutation in interferon regulatory factor 6 (Irf6), the homolog of the gene mutated in the human congenital disorders Van der Woude syndrome and popliteal pterygium syndrome, have a hyperproliferative epidermis that fails to undergo terminal differentiation, resulting in soft tissue fusions. We further demonstrate that mice that are compound heterozygotes for mutations in Irf6 and the gene encoding the cell cycle regulator protein stratifin (Sfn; also known as 14-3-3σ) show similar defects of keratinizing epithelia. Our results indicate that Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch and that Irf6 and Sfn interact genetically in this process.


Gene | 2013

MAP kinase signalling cascades and transcriptional regulation

Shen Hsi Yang; Andrew D. Sharrocks; Alan J. Whitmarsh

The MAP kinase (MAPK) signalling pathways play fundamental roles in a wide range of cellular processes and are often deregulated in disease states. One major mode of action for these pathways is in controlling gene expression, in particular through regulating transcription. In this review, we discuss recent significant advances in this area. In particular we focus on the mechanisms by which MAPKs are targeted to the nucleus and chromatin, and once there, how they impact on chromatin structure and subsequent gene regulation. We also discuss how systems biology approaches have contributed to our understanding of MAPK signaling networks, and also how the MAPK pathways intersect with other regulatory pathways in the nucleus. Finally, we summarise progress in studying the physiological functions of key MAPK transcriptional targets.


Biochemical Society Transactions | 2006

The JIP family of MAPK scaffold proteins

Alan J. Whitmarsh

The components of MAPK (mitogen-activated protein kinase) signalling pathways can assemble into complexes that are co-ordinated by regulatory proteins including scaffold proteins. There is increasing evidence that scaffold proteins (i) maintain signalling specificity and facilitate the activation of pathway components, (ii) localize pathway components to particular subcellular sites or to specific targets, and (iii) serve as a point of signal integration to allow regulation of MAPK pathways by other signalling events in the cell. One family of scaffold proteins that regulate signalling by stress-activated MAPKs are the JIPs [JNK (c-Jun N-terminal kinase)-interacting proteins]. JIP proteins have been demonstrated to form complexes with specific JNK and p38 MAPK signalling modules and to play important roles in brain development, neuronal trafficking, apoptosis, beta-cell function and insulin responses. Here, I briefly review our current understanding of the biochemical properties and physiological roles of JIP proteins.


Journal of Cell Science | 2009

Mouse ACF7 and Drosophila Short stop modulate filopodia formation and microtubule organisation during neuronal growth

Natalia Sánchez-Soriano; Mark A. Travis; Federico Dajas-Bailador; Catarina Gonçalves-Pimentel; Alan J. Whitmarsh; Andreas Prokop

Spectraplakins are large actin-microtubule linker molecules implicated in various processes, including gastrulation, wound healing, skin blistering and neuronal degeneration. Expression data for the mammalian spectraplakin ACF7 and genetic analyses of the Drosophila spectraplakin Short stop (Shot) suggest an important role during neurogenesis. Using three parallel neuronal culture systems we demonstrate that, like Shot, ACF7 is essential for axon extension and describe, for the first time, their subcellular functions during axonal growth. Firstly, both ACF7 and Shot regulate the organisation of neuronal microtubules, a role dependent on both the F-actin- and microtubule-binding domains. This role in microtubule organisation is probably the key mechanism underlying the roles of Shot and ACF7 in growth cone advance. Secondly, we found a novel role for ACF7 and Shot in regulating the actin cytoskeleton through their ability to control the formation of filopodia. This function in F-actin regulation requires EF-hand motifs and interaction with the translational regulator Krasavietz/eIF5C, indicating that the underlying mechanisms are completely different from those used to control microtubules. Our data provide the basis for the first mechanistic explanation for the role of Shot and ACF7 in the developing nervous system and demonstrate their ability to coordinate the organisation of both actin and microtubule networks during axonal growth.


Current Biology | 2008

The JIP1 Scaffold Protein Regulates Axonal Development in Cortical Neurons

Federico Dajas-Bailador; Emma V. Jones; Alan J. Whitmarsh

Summary The development of neuronal polarity is essential for the determination of neuron connectivity and for correct brain function. The c-Jun N-terminal kinase (JNK)-interacting protein-1 (JIP1) is highly expressed in neurons and has previously been characterized as a regulator of JNK signaling. JIP1 has been shown to localize to neurites in various neuronal models, but the functional significance of this localization is not fully understood [1–4]. JIP1 is also a cargo of the motor protein kinesin-1, which is important for axonal transport [2, 4]. Here we demonstrate that before primary cortical neurons become polarized, JIP1 specifically localizes to a single neurite and that after axonal specification, it accumulates in the emerging axon. JIP1 is necessary for normal axonal development and promotes axonal growth dependent upon its binding to kinesin-1 and via a newly described interaction with the c-Abl tyrosine kinase. JIP1 associates with and is phosphorylated by c-Abl, and the mutation of the c-Abl phosphorylation site on JIP1 abrogates its ability to promote axonal growth. JIP1 is therefore an important regulator of axonal development and is a key target of c-Abl-dependent pathways that control axonal growth.


Biochemical Journal | 2007

Regulation of p73-mediated apoptosis by c-Jun N-terminal kinase

Emma V. Jones; Mark J. Dickman; Alan J. Whitmarsh

The JNK (c-Jun N-terminal kinase)/mitogen-activated protein kinase signalling pathway is a major mediator of stress responses in cells, including the response to DNA damage. DNA damage also causes the stabilization and activation of p73, a member of the p53 family of transcription factors. p73, like p53, can mediate apoptosis by up-regulating the expression of pro-apoptotic genes, including Bax (Bcl2-associated X protein) and PUMA (p53 up-regulated modulator of apoptosis). Changes in p73 expression have been linked to tumour progression, particularly in neuroblastomas, whereas in tumours that feature inactivated p53 there is evidence that p73 may mediate the apoptotic response to chemotherapeutic agents. In the present study, we demonstrate a novel link between the JNK signalling pathway and p73. We use pharmacological and genetic approaches to show that JNK is required for p73-mediated apoptosis induced by the DNA damaging agent cisplatin. JNK forms a complex with p73 and phosphorylates it at several serine and threonine residues. The mutation of JNK phosphorylation sites in p73 abrogates cisplatin-induced stabilization of p73 protein, leading to a reduction in p73 transcriptional activity and reduced p73-mediated apoptosis. Our results demonstrate that the JNK pathway is an important regulator of DNA damage-induced apoptosis mediated by p73.


Methods in Enzymology | 2001

Analyzing JNK and p38 mitogen-activated protein kinase activity

Alan J. Whitmarsh; Roger J. Davis

Publisher Summary The JNK and p38 mitogen-activated protein (MAP) kinase groups are important for many physiological processes including cell growth, oncogenic transformation, cell differentiation, apoptosis, and the immune response. To understand the role of JNK and p38 MAP kinases in the cell, several protocols for measuring their activation state and protein kinase activity have been used. This chapter discusses many techniques for analyzing the activities of JNK and p38 mitogen-activated protein (MAP) kinases in vitro and in vivo . The choice of a particular technique depends on a number of factors, including whether (1) the overall activity or the activity of particular isoforms is to be measured, (2) the effect of inhibitors on JNK and p38 activation or protein kinase activity is being measured, or (3) the signaling events downstream or upstream of these MAP kinases are being analyzed. The development of improved antibodies to individual JNK and p38 isoforms as well as novel phosphorylation-specific antibodies that recognize the JNK and p38 target phosphorylation sites in substrates greatly aids the dissection of the roles of JNK and p38 family members in the cell. In addition, the promise of highly specific drugs that target particular JNK and p38 enzymes allows more complete understanding of these complex signaling pathways.


Trends in Biochemical Sciences | 2015

Mitochondrial Proteins Moonlighting in the Nucleus

Richard M. Monaghan; Alan J. Whitmarsh

Mitochondria function as cellular energy generators, producing the fuel required to drive biological processes. The response of cells to mitochondrial activity or dysfunction regulates their survival, growth, proliferation, and differentiation. Several proteins that contain mitochondrial-targeting sequences (MTS) also reside in the nucleus and there is increasing evidence that the nuclear translocation of mitochondrial proteins represents a novel pathway by which mitochondria signal their status to the cell. Here, we discuss the different mechanisms that control the dual mitochondrial and nuclear localisation of proteins and propose that these nuclear moonlighters represent a widespread regulatory circuit to maintain mitochondrial homeostasis.

Collaboration


Dive into the Alan J. Whitmarsh's collaboration.

Top Co-Authors

Avatar

Roger J. Davis

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julie Cavanagh

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emma V. Jones

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

Gino Poulin

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge