Network


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

Hotspot


Dive into the research topics where Helen Morrison is active.

Publication


Featured researches published by Helen Morrison.


Nature | 2006

Tumorigenic transformation by CPI-17 through inhibition of a merlin phosphatase

Hongchuan Jin; Tobias Sperka; Peter Herrlich; Helen Morrison

The tumour suppressor protein merlin (encoded by the neurofibromatosis type 2 gene NF2) is an important regulator of proliferation in many cell and tissue types. Merlin is activated by dephosphorylation at serine 518 (S518), which occurs on serum withdrawal or on cell–cell or cell–matrix contact. However, the relevant phosphatase that activates merlins tumour suppressor function is unknown. Here we identify this enzyme as the myosin phosphatase (MYPT-1–PP1δ). The cellular MYPT-1–PP1δ-specific inhibitor CPI-17 causes a loss of merlin function characterized by merlin phosphorylation, Ras activation and transformation. Constitutively active merlin (S518A) reverses CPI-17-induced transformation, showing that merlin is the decisive substrate of MYPT-1–PP1δ in tumour suppression. In addition we show that CPI-17 levels are raised in several human tumour cell lines and that the downregulation of CPI-17 induces merlin dephosphorylation, inhibits Ras activation and abolishes the transformed phenotype. MYPT-1–PP1δ and its substrate merlin are part of a previously undescribed tumour suppressor cascade that can be hindered in two ways, by mutation of the NF2 gene and by upregulation of the oncoprotein CPI-17.


Annals of the New York Academy of Sciences | 2006

CD44 Acts Both as a Growth‐ and Invasiveness‐Promoting Molecule and as a Tumor‐Suppressing Cofactor

Peter Herrlich; Helen Morrison; Jonathan P. Sleeman; Véronique Orian-Rousseau; Harald König; Susanne Weg-Remers; Helmut Ponta

Abstract: High‐molecular‐weight splice variants of the CD44 transmembrane protein family have been implicated in tumorigenesis and metastasis formation. By contrast, in certain tumors‐for example, Burkitts lymphoma, neuroblastomas, and prostate cancer‐loss of CD44 expression seems to accompany transformation. Here we describe two modes of action of CD44 proteins. They can bind growth factors and present them to their authentic high‐affinity receptors, and thus promote proliferation and invasiveness of cells. Under these conditions the CD44 proteins recruit ERM proteins‐for example, ezrin or moesin‐to their cytoplasmic tails, thereby producing links to the cytoskeleton. This mode of action could account for the tumor‐promoting action of CD44 proteins. The second mode of action of CD44 proteins comes into play when cells reach confluent growth conditions. Under specific conditions, binding of another ligand, the ECM component hyaluronate, leads to the activation and binding to the CD44 cytoplasmic tail of the tumor suppressor protein merlin. The activation of merlin confers growth arrest, so‐called contact inhibition. This function of CD44 proteins defines them as tumor suppressors. The type of action of CD44 on a given cell will depend on the isoform pattern of CD44 expressed, on the cellular equipment with ERM protein members, on the nature of the ECM, and on yet‐unknown conditions.


The EMBO Journal | 2005

Listeria monocytogenes exploits ERM protein functions to efficiently spread from cell to cell

Sascha Pust; Helen Morrison; Jürgen Wehland; Antonio S. Sechi; Peter Herrlich

Cell‐to‐cell spread is a fundamental step in the infection cycle of Listeria monocytogenes that strictly depends on the formation of bacteria‐induced protrusions. Since Listeria actin tails in the protrusions are tightly associated with the plasma membrane, we hypothesised that membrane–cytoskeleton linkers would be required for initiating and sustaining their formation and the subsequent cell‐to‐cell spread. We have found that ezrin, a member of the ezrin, radixin and moesin (ERM) family that functions as a key membrane–cytoskeleton linker, accumulates at Listeria protrusions. The ability of Listeria to induce protrusions and effectively spread between adjacent cells depends on the interaction of ERM proteins with both a membrane component such as CD44 and actin filaments. Interfering with either of these interactions or with ERM proteins phosphorylation not only reduces the number of protrusions but also alters their morphology, resulting in the formation of short and collapsed protrusions. As a consequence, Listeria cell‐to‐cell spread is severely impaired. Thus, ERM proteins are exploited by Listeria to escape the host immune response and to succeed in the development of the infection.


Nature Neuroscience | 2013

Merlin isoform 2 in neurofibromatosis type 2-associated polyneuropathy.

Alexander Schulz; Stephan L. Baader; Michiko Niwa-Kawakita; Marie Juliane Jung; Reinhard Bauer; Cynthia Garcia; Ansgar Zoch; Stephan Schacke; Christian Hagel; Victor-Felix Mautner; C. Oliver Hanemann; Xin-Peng Dun; David Parkinson; Joachim Weis; J. Michael Schröder; David H. Gutmann; Marco Giovannini; Helen Morrison

The autosomal dominant disorder neurofibromatosis type 2 (NF2) is a hereditary tumor syndrome caused by inactivation of the NF2 tumor suppressor gene, encoding merlin. Apart from tumors affecting the peripheral and central nervous systems, most NF2 patients develop peripheral neuropathies. This peripheral nerve disease can occur in the absence of nerve-damaging tumors, suggesting an etiology that is independent of gross tumor burden. We discovered that merlin isoform 2 (merlin-iso2) has a specific function in maintaining axonal integrity and propose that reduced axonal NF2 gene dosage leads to NF2-associated polyneuropathy. We identified a merlin-iso2–dependent complex that promotes activation of the GTPase RhoA, enabling downstream Rho-associated kinase to promote neurofilament heavy chain phosphorylation. Merlin-iso2–deficient mice exhibited impaired locomotor capacities, delayed sensory reactions and electrophysiological signs of axonal neuropathy. Sciatic nerves from these mice and sural nerve biopsies from NF2 patients revealed reduced phosphorylation of the neurofilament H subunit, decreased interfilament spacings and irregularly shaped axons.


Journal of Molecular Biology | 2009

Properties of an Ezrin Mutant Defective in F-actin Binding

Henri S. Saleh; Ulrike Merkel; Katja J. Geißler; Tobias Sperka; Antonio S. Sechi; Constanze Breithaupt; Helen Morrison

Ezrin, radixin and moesin are a family of proteins that provide a link between the plasma membrane and the cortical actin cytoskeleton. The regulated targeting of ezrin to the plasma membrane and its association with cortical F-actin are more than likely functions necessary for a number of cellular processes, such as cell adhesion, motility, morphogenesis and cell signalling. The interaction with F-actin was originally mapped to the last 34 residues of ezrin, which correspond to the last three helices (alphaB, alphaC and alphaD) of the C-terminal tail. We set out to identify and mutate the ezrin/F-actin binding site in order to pinpoint the role of F-actin interaction in morphological processes as well as signal transduction. We report here the generation of an ezrin mutant defective in F-actin binding. We identified four actin-binding residues, T576, K577, R579 and I580, that form a contiguous patch on the surface of the last helix, alphaD. Interestingly, mutagenesis of R579 also eliminated the interaction of band four-point one, ezrin, radixin, moesin homology domains (FERM) and the C-terminal tail domain, identifying a hotspot of the FERM/tail interaction. In vivo expression of the ezrin mutant defective in F-actin binding and FERM/tail interaction (R579A) altered the normal cell surface structure dramatically and inhibited cell migration. Further, we showed that ezrin/F-actin binding is required for the receptor tyrosine kinase signal transfer to the Ras/MAP kinase signalling pathway. Taken together, these observations highlight the importance of ezrin/F-actin function in the development of dynamic membrane/actin structures critical for cell shape and motility, as well as signal transduction.


The Journal of Neuroscience | 2010

Merlin Inhibits Neurite Outgrowth in the CNS

Alexander Schulz; Katja J. Geissler; Sujeet Kumar; Gregor Leichsenring; Helen Morrison; Stephan L. Baader

The neurofibromatosis type 2 gene product merlin is known to provoke gliogenic tumors as a result of its mutagenic loss. Merlins physiological anti-mitogenic function makes it unique among its ezrin–radixin–moesin (ERM) family members. Although ERM proteins and merlin are known to be expressed in glial cells of the peripheral nervous system and CNS, the neuronal expression pattern and function of merlin have been less well investigated. We report here expression of merlin in developing and mature neurons of the murine CNS. Within cerebellar Purkinje cells (PCs), merlin was localized in the soma, sprouting dendrites and axons. Merlin expression in PCs was high during the period of initial dendrite regression and declined during later phases of dendrite elongation. Consistently, merlin expression in vivo was increased in Engrailed-2-overexpressing PCs, which are characterized by a reduced dendritic extension. Furthermore, overexpression of merlin in dissociated cerebellar cultures and in neurogenic P19 cells caused a significant decline in neurite outgrowth, while, conversely, inhibition of merlin expression increased process formation. This effect was dependent on phosphorylation of serine 518 and involved the inactivation of the growth-promoting GTPase Rac. We thus provide evidence that merlin plays a pivotal role in controlling the neuronal wiring in the developing CNS.


American Journal of Medical Genetics Part A | 2010

What's new in neurofibromatosis? Proceedings from the 2009 NF conference: New frontiers

Joseph L. Kissil; Jaishri O. Blakeley; Rosalie E. Ferner; Susan M. Huson; Michel Kalamarides; Victor F. Mautner; Frank McCormick; Helen Morrison; Roger J. Packer; Vijaya Ramesh; Nancy Ratner; Katherine A. Rauen; David A. Stevenson; Kim Hunter-Schaedle; Kathryn N. North

The NF Conference is the largest annual gathering of researchers and clinicians focused on neurofibromatosis and has been convened by the Childrens Tumor Foundation for over 20 years. The 2009 NF Conference was held in Portland, Oregon from June 13 to June 16, 2009 and co‐chaired by Kathryn North from the University of Sydney and The Childrens Hospital at Westmead, Sydney, Australia; and Joseph Kissil from the Wistar Institute, Philadelphia. The Conference included 80 platform presentations in 9 sessions over 4 days; over 100 abstracts presented as posters; and three Keynote presentations. To date, there have been tremendous advances in basic research in the pathogenesis of neurofibromatosis, and more recently in progress toward identifying effective drug therapies and the commencement of neurofibromatosis clinical trials. The NF Conference attendees have significantly increased (doubling from 140 in 2005 to 280 attending in 2009) with a significant increase in attendance of physicians and clinical researchers. Correspondingly the NF Conference scope has expanded to include translational research, clinical trials and clinical management issues while retaining a core of basic research. These themes are reflected in the highlights from the 2009 NF Conference presented here.


Acta Neuropathologica | 2012

Neurofibromatosis 2011: A Report of the Children’s Tumor Foundation Annual Meeting

Michel Kalamarides; Maria T. Acosta; Dusica Babovic-Vuksanovic; Olli Carpén; Karen Cichowski; D. Gareth Evans; Filippo G. Giancotti; C. Oliver Hanemann; David A. Ingram; Alison C. Lloyd; Debra A. Mayes; Ludwine Messiaen; Helen Morrison; Kathryn N. North; Roger J. Packer; Duojia Pan; Anat Stemmer-Rachamimov; Meena Upadhyaya; David H. Viskochil; Margret R. Wallace; Kim Hunter-Schaedle; Nancy Ratner

The 2011 annual meeting of the Children’s Tumor Foundation, the annual gathering of the neurofibromatosis (NF) research and clinical communities, was attended by 330 participants who discussed integration of new signaling pathways into NF research, the appreciation for NF mutations in sporadic cancers, and an expanding pre-clinical and clinical agenda. NF1, NF2, and schwannomatosis collectively affect approximately 100,000 persons in US, and result from mutations in different genes. Benign tumors of NF1 (neurofibroma and optic pathway glioma) and NF2 (schwannoma, ependymoma, and meningioma) and schwannomatosis (schwannoma) can cause significant morbidity, and there are no proven drug treatments for any form of NF. Each disorder is associated with additional manifestations causing morbidity. The research presentations described in this review covered basic science, preclinical testing, and results from clinical trials, and demonstrate the remarkable strides being taken toward understanding of and progress toward treatments for these disorders based on the close interaction among scientists and clinicians.


Journal of Cell Biology | 2017

Merlin controls the repair capacity of Schwann cells after injury by regulating Hippo/YAP activity

Thomas Mindos; Xin-Peng Dun; Katherine North; Robin D. S. Doddrell; Alexander Schulz; Philip Edwards; James A. Russell; Bethany Gray; Sheridan L. Roberts; Aditya Shivane; Georgina Mortimer; Melissa Pirie; Nailing Zhang; Duojia Pan; Helen Morrison; David Parkinson

Loss of the Merlin tumor suppressor and activation of the Hippo signaling pathway play major roles in the control of cell proliferation and tumorigenesis. We have identified completely novel roles for Merlin and the Hippo pathway effector Yes-associated protein (YAP) in the control of Schwann cell (SC) plasticity and peripheral nerve repair after injury. Injury to the peripheral nervous system (PNS) causes a dramatic shift in SC molecular phenotype and the generation of repair-competent SCs, which direct functional repair. We find that loss of Merlin in these cells causes a catastrophic failure of axonal regeneration and remyelination in the PNS. This effect is mediated by activation of YAP expression in Merlin-null SCs, and loss of YAP restores axonal regrowth and functional repair. This work identifies new mechanisms that control the regenerative potential of SCs and gives new insight into understanding the correct control of functional nerve repair in the PNS.


Brain | 2014

Neuronal merlin influences ERBB2 receptor expression on Schwann cells through neuregulin 1 type III signalling

Alexander Schulz; Anna Kyselyova; Stephan L. Baader; Marie Juliane Jung; Ansgar Zoch; Victor-Felix Mautner; Christian Hagel; Helen Morrison

Merlin mutations in Neurofibromatosis type 2 cause tumorigenic transformation of Schwann cells, leading to schwannoma development. Schulz et al. show that loss of neuronally expressed merlin alone increases the susceptibility of adjacent Schwann cells to mitogenic signals through the Neuregulin1-ErbB2/3 pathway.

Collaboration


Dive into the Helen Morrison's collaboration.

Top Co-Authors

Avatar

Alexander Schulz

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter Herrlich

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David H. Gutmann

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

David Parkinson

Plymouth State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge