Network


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

Hotspot


Dive into the research topics where Thomas Worzfeld is active.

Publication


Featured researches published by Thomas Worzfeld.


Journal of Biological Chemistry | 2008

ErbB-2 and Met Reciprocally Regulate Cellular Signaling via Plexin-B1

Jakub M. Swiercz; Thomas Worzfeld; Stefan Offermanns

Sema4D-induced activation of plexin-B1 has been reported to evoke different and sometimes opposing cellular responses. The mechanisms underlying the versatility of plexin-B1-mediated effects are not clear. Plexin-B1 can associate with the receptor tyrosine kinases ErbB-2 and Met. Here we show that Sema4D-induced activation and inactivation of RhoA require ErbB-2 and Met, respectively. In breast carcinoma cells, Sema4D can have pro- and anti-migratory effects depending on the presence of ErbB-2 and Met, and the exchange of the two receptor tyrosine kinases is sufficient to convert the cellular response to Sema4D from pro- to anti-migratory and vice versa. This work identifies a novel mechanism by which plexin-mediated signaling can be regulated and explains how Sema4D can exert different biological activities through the differential association of its receptor with ErbB-2 and Met.


Trends in Pharmacological Sciences | 2008

G12/G13-mediated signalling in mammalian physiology and disease

Thomas Worzfeld; Nina Wettschureck; Stefan Offermanns

The human genome encodes hundreds of G-protein-coupled receptors. Their intracellular effects, however, are mediated by only four families of heterotrimeric G proteins: G(s), G(i)/G(o), G(q)/G(11) and G(12)/G(13). Progress in the knowledge about the G(12)/G(13) family has somewhat lagged behind because their downstream effectors remained unknown for several years, and tools to specifically interfere with G(12)/G(13)-mediated signalling were, therefore, missing. However, with the identification of G(12)/G(13)-regulated signalling pathways and the recent application of new techniques, such as conditional gene inactivation, RNA interference or expression of inhibitory proteins, new insights into the in vivo functions of this G-protein family have been gained. It has become clear that this pathway regulates cellular proliferation, movement and morphology in many different organs and that it is centrally involved in various diseases including cancer and cardiovascular disorders. Here, we focus on recent progress made in the analyses of the in vivo functions of mammalian G(12)/G(13)-mediated signalling.


Nature Reviews Drug Discovery | 2014

Semaphorins and plexins as therapeutic targets

Thomas Worzfeld; Stefan Offermanns

Semaphorins are membrane-bound or diffusible factors that regulate key cellular functions and are involved in cell–cell communication. Most of the effects of semaphorins are mediated by plexins. Work over the past decade has revealed crucial functions of the semaphorin–plexin system in mammalian physiology. It has also become clear that semaphorins and plexins have important roles in many pathophysiological processes, including cancer, immunological diseases and bone disorders, and that they represent novel targets for drugs to prevent or treat various diseases. This Review summarizes the functions of the mammalian semaphorin–plexin system as well as its role in diseases and discusses emerging strategies to pharmacologically target semaphorin–plexin signalling.


European Journal of Neuroscience | 2004

Plexin-B family members demonstrate non-redundant expression patterns in the developing mouse nervous system: an anatomical basis for morphogenetic effects of Sema4D during development

Thomas Worzfeld; Andreas W. Püschel; Stefan Offermanns; Rohini Kuner

Semaphorins and their receptors play important roles in patterning the connectivity of the developing nervous system and recent data suggest that members of the plexin‐B family of semaphorin receptors may be involved in axonal guidance. Here we show that the mRNAs of the three plexin‐B genes, plxnb1, plxnb2 and plxnb3 (plexin‐B1, plexin‐B2 and plexin‐B3), respectively, are expressed in highly specific and non‐redundant patterns in peripheral and central components of the nervous system over defined periods during murine development. Whereas plexin‐B1 and plexin‐B2 are strongly expressed in the neuroepithelium and developing neurons, plexin‐B3 mRNA is selectively localized to the white matter. Moreover, plexin‐B1 and its ligand Sema4D are expressed in complementary patterns in several regions such as the developing neopallial cortex, the dorsal root ganglia and the spinal cord over embryonic stages. The Sema4d gene demonstrates a dramatic switch from prenatal expression in neuronal populations to a postnatal expression in oligodendrocytes. In contrast to its collapsing activity on growth cones of embryonic retinal ganglion cells and hippocampal neurons, soluble Sema4D enhances axonal outgrowth in embryonic cortical explants cultured in collagen matrices. Thus, plexin‐B family members and Sema4D are likely to play complex and non‐redundant roles during the development of the nervous system.


The Journal of Neuroscience | 2007

Plexin-B2, But Not Plexin-B1, Critically Modulates Neuronal Migration and Patterning of the Developing Nervous System In Vivo

Suhua Deng; Alexandra Hirschberg; Thomas Worzfeld; Junia Y. Penachioni; Alexander Korostylev; Jakub M. Swiercz; Peter Vodrazka; Olivier Mauti; Esther T. Stoeckli; Luca Tamagnone; Stefan Offermanns; Rohini Kuner

Semaphorins and their receptors, plexins, have emerged as important cellular cues regulating key developmental processes. B-type plexins directly regulate the actin cytoskeleton in a variety of cell types. Recently, B-type plexins have been shown to be expressed in striking patterns in the nervous system over critical developmental windows. However, in contrast to the well characterized plexin-A family, the functional role of plexin-B proteins in neural development and organogenesis in vertebrates in vivo is not known. Here, we have elucidated the functional contribution of the two neuronally expressed plexin-B proteins, Plexin-B1 or Plexin-B2, toward the development of the peripheral nervous system and the CNS by generating and analyzing constitutive knock-out mice. The development of the nervous system was found to be normal in mice lacking Plexin-B1, whereas mice lacking Plexin-B2 demonstrated defects in closure of the neural tube and a conspicuous disorganization of the embryonic brain. After analyzing mutant mice, which bypassed neural tube defects, we observed a key requirement for Plexin-B2 in proliferation and migration of granule cell precursors in the developing dentate gyrus, olfactory bulb, and cerebellum. Furthermore, we identified semaphorin 4C as a high-affinity ligand for Plexin-B2 in binding and functional assays. Semaphorin 4C stimulated activation of ErbB-2 and RhoA via Plexin-B2 and enhanced proliferation and migration of granule cell precursors. Semaphorin 4C-induced proliferation of ventricular zone neuroblasts was abrogated in mice lacking Plexin-B2. These genetic and functional analyses reveal a key requirement for Plexin-B2, but not Plexin-B1, in patterning of the vertebrate nervous system in vivo.


Journal of Clinical Investigation | 2012

ErbB-2 signals through Plexin-B1 to promote breast cancer metastasis

Thomas Worzfeld; Jakub M. Swiercz; Mario Looso; Beate K. Straub; Kishor K. Sivaraj; Stefan Offermanns

Diagnosis of metastatic breast cancer is associated with a very poor prognosis. New therapeutic targets are urgently needed, but their development is hampered by a lack of understanding of the mechanisms leading to tumor metastasis. Exemplifying this is the fact that the approximately 30% of all breast cancers overexpressing the receptor tyrosine kinase ErbB-2 are characterized by high metastatic potential and poor prognosis, but the signaling events downstream of ErbB-2 that drive cancer cell invasion and metastasis remain incompletely understood. Here we show that overexpression of ErbB-2 in human breast cancer cell lines leads to phosphorylation and activation of the semaphorin receptor Plexin-B1. This was required for ErbB-2-dependent activation of the pro-metastatic small GTPases RhoA and RhoC and promoted invasive behavior of human breast cancer cells. In a mouse model of ErbB-2-overexpressing breast cancer, ablation of the gene encoding Plexin-B1 strongly reduced the occurrence of metastases. Moreover, in human patients with ErbB-2-overexpressing breast cancer, low levels of Plexin-B1 expression correlated with good prognosis. Our data suggest that Plexin-B1 represents a new candidate therapeutic target for treating patients with ErbB-2-positive breast cancer.


Development | 2008

A functional role for semaphorin 4D/plexin B1 interactions in epithelial branching morphogenesis during organogenesis

Alexander Korostylev; Thomas Worzfeld; Suhua Deng; Roland H. Friedel; Jakub M. Swiercz; Peter Vodrazka; Viola Maier; Alexandra Hirschberg; Yoshiharu Ohoka; Shinobu Inagaki; Stefan Offermanns; Rohini Kuner

Semaphorins and their receptors, plexins, carry out important functions during development and disease. In contrast to the well-characterized plexin A family, however, very little is known about the functional relevance of B-type plexins in organogenesis, particularly outside the nervous system. Here, we demonstrate that plexin B1 and its ligand Sema4d are selectively expressed in epithelial and mesenchymal compartments during key steps in the genesis of some organs. This selective expression suggests a role in epithelial-mesenchymal interactions. Importantly, using the developing metanephros as a model system, we have observed that endogenously expressed and exogenously supplemented Sema4d inhibits branching morphogenesis during early stages of development of the ureteric collecting duct system. Our results further suggest that the RhoA-ROCK pathway, which is activated downstream of plexin B1, mediates these inhibitory morphogenetic effects of Sema4d and suppresses branch-promoting signalling effectors of the plexin B1 signalling complex. Finally, mice that lack plexin B1 show early anomalies in kidney development in vivo. These results identify a novel function for plexin B1 as a negative regulator of branching morphogenesis during kidney development, and suggest that the Sema4d-plexin B1 ligand-receptor pair contributes to epithelial-mesenchymal interactions during organogenesis via modulation of RhoA signalling.


Molecular and Cellular Biology | 2010

Gene Deletion Mutants Reveal a Role for Semaphorin Receptors of the Plexin-B Family in Mechanisms Underlying Corticogenesis

Alexandra Hirschberg; Suhua Deng; Alexander Korostylev; Eszter Paldy; Marcos R. Costa; Thomas Worzfeld; Peter Vodrazka; Andrea Wizenmann; Magdalena Götz; Stefan Offermanns; Rohini Kuner

ABSTRACT Semaphorins and their receptors, plexins, are emerging as key regulators of various aspects of neural and nonneural development. Semaphorin 4D (Sema4D) and B-type plexins demonstrate distinct expression patterns over critical time windows during the development of the murine neocortex. Here, analysis of mice genetically lacking plexin-B1 or plexin-B2 revealed the significance of Sema4D-plexin-B signaling in cortical development. Deficiency of plexin-B2 resulted in abnormal cortical layering and defective migration and differentiation of several subtypes of cortical neurons, including Cajal-Retzius cells, GABAergic interneurons, and principal cells in vivo. In contrast, a lack of plexin-B1 did not impact on cortical development in vivo. In various ex vivo assays on embryonic forebrain, Sema4D enhanced the radial and tangential migration of developing neurons in a plexin-B2-dependent manner. These results suggest that Sema4D-plexin-B2 interactions regulate mechanisms underlying cell specification, differentiation, and migration during corticogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Genetic dissection of plexin signaling in vivo

Thomas Worzfeld; Jakub M. Swiercz; Aycan Sentürk; Berit Genz; Alexander Korostylev; Suhua Deng; Jingjing Xia; Mikio Hoshino; Jonathan A. Epstein; Andrew M. Chan; Brigitte Vollmar; Amparo Acker-Palmer; Rohini Kuner; Stefan Offermanns

Significance Plexins, a family of transmembrane receptors for semaphorins, control diverse biological processes during mouse development. However, it is largely unknown through which signaling pathways they exert their functions in vivo. Using an allelic series of transgenic mice, we show that the GTPase activating protein domain of plexins constitutes their key signaling module during development, which is required for proper formation of the nervous, cardiovascular, and skeletal system. In contrast, development of the liver vasculature specifically depends on the activation of the small GTPase RhoA by the plexin family member Plexin-B2. This study uncovers the in vivo context-dependence and functional specificity of individual plexin-mediated signaling pathways during mouse development. Mammalian plexins constitute a family of transmembrane receptors for semaphorins and represent critical regulators of various processes during development of the nervous, cardiovascular, skeletal, and renal system. In vitro studies have shown that plexins exert their effects via an intracellular R-Ras/M-Ras GTPase-activating protein (GAP) domain or by activation of RhoA through interaction with Rho guanine nucleotide exchange factor proteins. However, which of these signaling pathways are relevant for plexin functions in vivo is largely unknown. Using an allelic series of transgenic mice, we show that the GAP domain of plexins constitutes their key signaling module during development. Mice in which endogenous Plexin-B2 or Plexin-D1 is replaced by transgenic versions harboring mutations in the GAP domain recapitulate the phenotypes of the respective null mutants in the developing nervous, vascular, and skeletal system. We further provide genetic evidence that, unexpectedly, the GAP domain-mediated developmental functions of plexins are not brought about via R-Ras and M-Ras inactivation. In contrast to the GAP domain mutants, Plexin-B2 transgenic mice defective in Rho guanine nucleotide exchange factor binding are viable and fertile but exhibit abnormal development of the liver vasculature. Our genetic analyses uncover the in vivo context-dependence and functional specificity of individual plexin-mediated signaling pathways during development.


Molecular and Cellular Biology | 2009

Semaphorin 4D Signaling Requires the Recruitment of Phospholipase Cγ into the Plexin-B1 Receptor Complex

Jakub M. Swiercz; Thomas Worzfeld; Stefan Offermanns

ABSTRACT The semaphorin 4D (Sema4D) receptor plexin-B1 constitutively interacts with particular Rho guanine nucleotide exchange factors (RhoGEFs) and thereby mediates Sema4D-induced RhoA activation, a process which involves the tyrosine phosphorylation of plexin-B1 by ErbB-2. It is, however, unknown how plexin-B1 phosphorylation regulates RhoGEF activity. We show here that activation of plexin-B1 by Sema4D and its subsequent tyrosine phosphorylation creates docking sites for the SH2 domains of phospholipase Cγ (PLCγ). PLCγ is thereby recruited into the plexin-B1 receptor complex and via its SH3 domain activates the Rho guanine nucleotide exchange factor PDZ-RhoGEF. PLCγ-dependent RhoGEF activation is independent of its lipase activity. The recruitment of PLCγ has no effect on the R-Ras GTPase-activating protein activity of plexin-B1 but is required for Sema4D-induced axonal growth cone collapse as well as for the promigratory effects of Sema4D on cancer cells. These data demonstrate a novel nonenzymatic function of PLCγ as an important mechanism of plexin-mediated signaling which links tyrosine phosphorylation of plexin-B1 to the regulation of a RhoGEF protein and downstream cellular processes.

Collaboration


Dive into the Thomas Worzfeld's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge