Network


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

Hotspot


Dive into the research topics where Phebe S. Wulf is active.

Publication


Featured researches published by Phebe S. Wulf.


Neuron | 2009

Dynamic Microtubules Regulate Dendritic Spine Morphology and Synaptic Plasticity

Jacek Jaworski; Lukas C. Kapitein; Susana Montenegro Gouveia; Bjorn Dortland; Phebe S. Wulf; Ilya Grigoriev; Paola Camera; Samantha A. Spangler; Paola Di Stefano; Jeroen Demmers; Harm J. Krugers; Paola Defilippi; Anna Akhmanova; Casper C. Hoogenraad

Dendritic spines are the major sites of excitatory synaptic input, and their morphological changes have been linked to learning and memory processes. Here, we report that growing microtubule plus ends decorated by the microtubule tip-tracking protein EB3 enter spines and can modulate spine morphology. We describe p140Cap/SNIP, a regulator of Src tyrosine kinase, as an EB3 interacting partner that is predominantly localized to spines and enriched in the postsynaptic density. Inhibition of microtubule dynamics, or knockdown of either EB3 or p140Cap, modulates spine shape via regulation of the actin cytoskeleton. Fluorescence recovery after photobleaching revealed that EB3-binding is required for p140Cap accumulation within spines. In addition, we found that p140Cap interacts with Src substrate and F-actin-binding protein cortactin. We propose that EB3-labeled growing microtubule ends regulate the localization of p140Cap, control cortactin function, and modulate actin dynamics within dendritic spines, thus linking dynamic microtubules to spine changes and synaptic plasticity.


Nature Cell Biology | 2002

Bicaudal-D regulates COPI-independent Golgi-ER transport by recruiting the dynein-dynactin motor complex

Theodoros Matanis; Anna Akhmanova; Phebe S. Wulf; Elaine Del Nery; Thomas Weide; Tatiana Stepanova; Niels Galjart; Frank Grosveld; Bruno Goud; Chris I. De Zeeuw; Angelika Barnekow; Casper C. Hoogenraad

The small GTPase Rab6a is involved in the regulation of membrane traffic from the Golgi apparatus towards the endoplasmic reticulum (ER) in a coat complex coatomer protein I (COPI)-independent pathway. Here, we used a yeast two-hybrid approach to identify binding partners of Rab6a. In particular, we identified the dynein–dynactin-binding protein Bicaudal-D1 (BICD1), one of the two mammalian homologues of Drosophila Bicaudal-D. BICD1 and BICD2 colocalize with Rab6a on the trans-Golgi network (TGN) and on cytoplasmic vesicles, and associate with Golgi membranes in a Rab6-dependent manner. Overexpression of BICD1 enhances the recruitment of dynein–dynactin to Rab6a-containing vesicles. Conversely, overexpression of the carboxy-terminal domain of BICD, which can interact with Rab6a but not with cytoplasmic dynein, inhibits microtubule minus-end-directed movement of green fluorescent protein (GFP)–Rab6a vesicles and induces an accumulation of Rab6a and COPI-independent ER cargo in peripheral structures. These data suggest that coordinated action between Rab6a, BICD and the dynein–dynactin complex controls COPI-independent Golgi–ER transport.


Current Biology | 2010

Mixed microtubules steer dynein-driven cargo transport into dendrites.

Lukas C. Kapitein; Max A. Schlager; Marijn Kuijpers; Phebe S. Wulf; Myrrhe van Spronsen; F. C. MacKintosh; Casper C. Hoogenraad

BACKGROUND To establish and maintain their polarized morphology, neurons employ active transport driven by molecular motors to sort cargo between axons and dendrites. However, the basic traffic rules governing polarized transport on neuronal microtubule arrays are unclear. RESULTS Here we show that the microtubule minus-end-directed motor dynein is required for the polarized targeting of dendrite-specific cargo, such as AMPA receptors. To directly examine how dynein motors contribute to polarized dendritic transport, we established a trafficking assay in hippocampal neurons to selectively probe specific motor protein activity. This revealed that, unlike kinesins, dynein motors drive cargo selectively into dendrites, governed by their mixed microtubule array. Moreover, axon-specific cargos, such as presynaptic vesicle protein synaptophysin, are redirected to dendrites by coupling to dynein motors. Quantitative modeling demonstrated that bidirectional dynein-driven transport on mixed microtubules provides an efficient mechanism to establish a stable density of continuously renewing vesicles in dendrites. CONCLUSIONS These results demonstrate a powerful approach to study specific motor protein activity inside living cells and imply a key role for dynein in dendritic transport. We propose that dynein establishes the initial sorting of dendritic cargo and additional motor proteins assist in subsequent delivery.


Neuron | 2013

TRAK/Milton motor-adaptor proteins steer mitochondrial trafficking to axons and dendrites.

Myrrhe van Spronsen; Marina Mikhaylova; Joanna Lipka; Max A. Schlager; Dave J. van den Heuvel; Marijn Kuijpers; Phebe S. Wulf; Nanda Keijzer; Jeroen Demmers; Lukas C. Kapitein; Dick Jaarsma; Hans C. Gerritsen; Anna Akhmanova; Casper C. Hoogenraad

In neurons, the distinct molecular composition of axons and dendrites is established through polarized targeting mechanisms, but it is currently unclear how nonpolarized cargoes, such as mitochondria, become uniformly distributed over these specialized neuronal compartments. Here, we show that TRAK family adaptor proteins, TRAK1 and TRAK2, which link mitochondria to microtubule-based motors, are required for axonal and dendritic mitochondrial motility and utilize different transport machineries to steer mitochondria into axons and dendrites. TRAK1 binds to both kinesin-1 and dynein/dynactin, is prominently localized in axons, and is needed for normal axon outgrowth, whereas TRAK2 predominantly interacts with dynein/dynactin, is more abundantly present in dendrites, and is required for dendritic development. These functional differences follow from their distinct conformations: TRAK2 preferentially adopts a head-to-tail interaction, which interferes with kinesin-1 binding and axonal transport. Our study demonstrates how the molecular interplay between bidirectional adaptor proteins and distinct microtubule-based motors drives polarized mitochondrial transport.


The EMBO Journal | 2003

Bicaudal D induces selective dynein‐mediated microtubule minus end‐directed transport

Casper C. Hoogenraad; Phebe S. Wulf; Natalia Schiefermeier; Tatiana Stepanova; Niels Galjart; J. Victor Small; Frank Grosveld; Chris I. De Zeeuw; Anna Akhmanova

Bicaudal D is an evolutionarily conserved protein, which is involved in dynein‐mediated motility both in Drosophila and in mammals. Here we report that the N–terminal portion of human Bicaudal D2 (BICD2) is capable of inducing microtubule minus end‐directed movement independently of the molecular context. This characteristic offers a new tool to exploit the relocalization of different cellular components by using appropriate targeting motifs. Here, we use the BICD2 N–terminal domain as a chimera with mitochondria and peroxisome‐anchoring sequences to demonstrate the rapid dynein‐mediated transport of selected organelles. Surprisingly, unlike other cytoplasmic dynein‐mediated processes, this transport shows very low sensitivity to overexpression of the dynactin subunit dynamitin. The dynein‐recruiting activity of the BICD2 N–terminal domain is reduced within the full‐length molecule, indicating that the C–terminal part of the protein might regulate the interaction between BICD2 and the motor complex. Our findings provide a novel model system for dissection of the molecular mechanism of dynein motility.


Neuron | 2014

Microtubule Minus-End Binding Protein CAMSAP2 Controls Axon Specification and Dendrite Development

Kah Wai Yau; Sam F.B. van Beuningen; Inês Cunha-Ferreira; Bas M. C. Cloin; Eljo Y. van Battum; Lena Will; Philipp Schätzle; Roderick P. Tas; Jaap van Krugten; Eugene A. Katrukha; Kai Jiang; Phebe S. Wulf; Marina Mikhaylova; Martin Harterink; R. Jeroen Pasterkamp; Anna Akhmanova; Lukas C. Kapitein; Casper C. Hoogenraad

In neurons, most microtubules are not associated with a central microtubule-organizing center (MTOC), and therefore, both the minus and plus-ends of these non-centrosomal microtubules are found throughout the cell. Microtubule plus-ends are well established as dynamic regulatory sites in numerous processes, but the role of microtubule minus-ends has remained poorly understood. Using live-cell imaging, high-resolution microscopy, and laser-based microsurgery techniques, we show that the CAMSAP/Nezha/Patronin family protein CAMSAP2 specifically localizes to non-centrosomal microtubule minus-ends and is required for proper microtubule organization in neurons. CAMSAP2 stabilizes non-centrosomal microtubules and is required for neuronal polarity, axon specification, and dendritic branch formation in vitro and in vivo. Furthermore, we found that non-centrosomal microtubules in dendrites are largely generated by γ-Tubulin-dependent nucleation. We propose a two-step model in which γ-Tubulin initiates the formation of non-centrosomal microtubules and CAMSAP2 stabilizes the free microtubule minus-ends in order to control neuronal polarity and development.


The EMBO Journal | 2010

Pericentrosomal targeting of Rab6 secretory vesicles by Bicaudal-D-related protein 1 (BICDR-1) regulates neuritogenesis

Max A. Schlager; Lukas C. Kapitein; Ilya Grigoriev; Grzegorz Burzynski; Phebe S. Wulf; Nanda Keijzer; Esther de Graaff; Mitsunori Fukuda; Iain T. Shepherd; Anna Akhmanova; Casper C. Hoogenraad

Membrane and secretory trafficking are essential for proper neuronal development. However, the molecular mechanisms that organize secretory trafficking are poorly understood. Here, we identify Bicaudal‐D‐related protein 1 (BICDR‐1) as an effector of the small GTPase Rab6 and key component of the molecular machinery that controls secretory vesicle transport in developing neurons. BICDR‐1 interacts with kinesin motor Kif1C, the dynein/dynactin retrograde motor complex, regulates the pericentrosomal localization of Rab6‐positive secretory vesicles and is required for neural development in zebrafish. BICDR‐1 expression is high during early neuronal development and strongly declines during neurite outgrowth. In young neurons, BICDR‐1 accumulates Rab6 secretory vesicles around the centrosome, restricts anterograde secretory transport and inhibits neuritogenesis. Later during development, BICDR‐1 expression is strongly reduced, which permits anterograde secretory transport required for neurite outgrowth. These results indicate an important role for BICDR‐1 as temporal regulator of secretory trafficking during the early phase of neuronal differentiation.


Developmental Cell | 2013

CFEOM1-Associated Kinesin KIF21A Is a Cortical Microtubule Growth Inhibitor

Babet van der Vaart; Wilhelmina E. van Riel; Harinath Doodhi; Josta T. Kevenaar; Eugene A. Katrukha; Laura F. Gumy; Benjamin P. Bouchet; Ilya Grigoriev; Samantha A. Spangler; Ka Lou Yu; Phebe S. Wulf; Jingchao Wu; Gideon Lansbergen; Eljo Y. van Battum; R. Jeroen Pasterkamp; Yuko Mimori-Kiyosue; Jeroen Demmers; Natacha Olieric; Ivan V. Maly; Casper C. Hoogenraad; Anna Akhmanova

Mechanisms controlling microtubule dynamics at the cell cortex play a crucial role in cell morphogenesis and neuronal development. Here, we identified kinesin-4 KIF21A as an inhibitor of microtubule growth at the cell cortex. In vitro, KIF21A suppresses microtubule growth and inhibits catastrophes. In cells, KIF21A restricts microtubule growth and participates in organizing microtubule arrays at the cell edge. KIF21A is recruited to the cortex by KANK1, which coclusters with liprin-α1/β1 and the components of the LL5β-containing cortical microtubule attachment complexes. Mutations in KIF21A have been linked to congenital fibrosis of the extraocular muscles type 1 (CFEOM1), a dominant disorder associated with neurodevelopmental defects. CFEOM1-associated mutations relieve autoinhibition of the KIF21A motor, and this results in enhanced KIF21A accumulation in axonal growth cones, aberrant axon morphology, and reduced responsiveness to inhibitory cues. Our study provides mechanistic insight into cortical microtubule regulation and suggests that altered microtubule dynamics contribute to CFEOM1 pathogenesis.


The Journal of Neuroscience | 2011

NMDA Receptor Activation Suppresses Microtubule Growth and Spine Entry

Lukas C. Kapitein; Kah Wai Yau; Susana Montenegro Gouveia; Wouter A. van der Zwan; Phebe S. Wulf; Nanda Keijzer; Jeroen Demmers; Jacek Jaworski; Anna Akhmanova; Casper C. Hoogenraad

Dynamic microtubules are important to maintain neuronal morphology and function, but whether neuronal activity affects the organization of dynamic microtubules is unknown. Here, we show that a protocol to induce NMDA-dependent long-term depression (LTD) rapidly attenuates microtubule dynamics in primary rat hippocampal neurons, removing the microtubule-binding protein EB3 from the growing microtubule plus-ends in dendrites. This effect requires the entry of calcium and is mediated by activation of NR2B-containing NMDA-type glutamate receptor. The rapid NMDA effect is followed by a second, more prolonged response, during which EB3 accumulates along MAP2-positive microtubule bundles in the dendritic shaft. MAP2 is both required and sufficient for this activity-dependent redistribution of EB3. Importantly, NMDA receptor activation suppresses microtubule entry in dendritic spines, whereas overexpression of EB3-GFP prevents NMDA-induced spine shrinkage. These results suggest that short-lasting and long-lasting changes in dendritic microtubule dynamics are important determinants for NMDA-induced LTD.


PLOS Biology | 2010

Neuron Specific Rab4 Effector GRASP-1 Coordinates Membrane Specialization and Maturation of Recycling Endosomes

Casper C. Hoogenraad; Ioana Popa; Kensuke Futai; Emma Sanchez-Martinez; Phebe S. Wulf; Thijs van Vlijmen; Bjorn Dortland; Viola Oorschot; Roland Govers; Maria Monti; Albert J. R. Heck; Morgan Sheng; Judith Klumperman; Holger Rehmann; Dick Jaarsma; Lukas C. Kapitein; Peter van der Sluijs

The neuronal protein GRASP-1 is shown to be a key molecule controlling endosomal trafficking and thereby regulating synapse integrity and synaptic plasticity.

Collaboration


Dive into the Phebe S. Wulf's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Max A. Schlager

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Nanda Keijzer

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Chris I. De Zeeuw

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Dick Jaarsma

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Jeroen Demmers

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Frank Grosveld

Erasmus University Rotterdam

View shared research outputs
Researchain Logo
Decentralizing Knowledge