Top Banner
Neuron 52, 293–305, October 19, 2006 ª2006 Elsevier Inc. DOI 10.1016/j.neuron.2006.08.018 Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway Katrin Deinhardt, 1,5 Sara Salinas, 1,5 Carole Verastegui, 1 Rose Watson, 2 Daniel Worth, 1 Sarah Hanrahan, 3 Cecilia Bucci, 4 and Giampietro Schiavo 1, * 1 Molecular NeuroPathobiology Laboratory 2 Electron Microscopy Laboratory 3 Protein Analysis Laboratory Cancer Research UK London Research Institute 44 Lincoln’s Inn Fields London, WC2A 3PX United Kingdom 4 Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali Universita ` di Lecce, via Provinciale Monteroni 73100 Lecce Italy Summary Vesicular pathways coupling the neuromuscular junc- tion with the motor neuron soma are essential for neu- ronal function and survival. To characterize the organ- elles responsible for this long-distance crosstalk, we developed a purification strategy based on a fragment of tetanus neurotoxin (TeNT H C ) conjugated to para- magnetic beads. This approach enabled us to identify, among other factors, the small GTPase Rab7 as a func- tional marker of a specific pool of axonal retrograde carriers, which transport neurotrophins and their receptors. Furthermore, Rab5 is essential for an early step in TeNT H C sorting but is absent from axonally transported vesicles. Our data demonstrate that TeNT H C uses a retrograde transport pathway shared with p75 NTR , TrkB, and BDNF, which is strictly depen- dent on the activities of both Rab5 and Rab7. There- fore, Rab7 plays an essential role in axonal retrograde transport by controlling a vesicular compartment implicated in neurotrophin traffic. Introduction Neurons represent the ultimate example of polarized cells. The maintenance of their unique morphology, which is based on axonal and dendritic arborization, is necessary for neuronal function and requires a complex network of trafficking pathways to allow efficient com- munication over long distances (Goldstein and Yang, 2000). Axonal transport constitutes the backbone of long-range crosstalk in neurons and is essential for their survival and differentiation. One of the most convincing demonstrations of the crucial role of axonal transport in neuronal homeostasis is the causal link between defects in this process and neurodegeneration (Holzbaur, 2004). In particular, key components of retrograde transport routes, which are responsible for the targeting of neuro- trophins and their survival signals to the soma, have been associated with motor neuron disease in humans and mice (Hafezparast et al., 2003; Puls et al., 2003). Despite the importance of these axonal compartments, little is known about their nature and the machinery reg- ulating retrograde transport in neurons. To date, few attempts to purify axonal retrograde transport compartments have been carried out. Re- cently, Delcroix et al. have provided a preliminary char- acterization of Trk-positive signaling endosomes, which contain activated Erk1/2, p38, and Akt, using a sciatic nerve chamber approach (Delcroix et al., 2003). In addi- tion, several signaling proteins triggered by nerve injury and undergoing retrograde transport have been identi- fied by a proteomic analysis in Lymnaea (Perlson et al., 2004). Here, we sought to characterize the axonal retro- grade compartment of spinal cord motor neurons (MN) by setting up a purification protocol based on paramag- netic iron beads (Fe beads) covalently bound to a frag- ment of tetanus neurotoxin (TeNT H C ). TeNT H C binds with high affinity to MN and is endocytosed by a specific clathrin-dependent pathway linked to a retrograde route to the central nervous system (Deinhardt et al., 2006; Roux et al., 2005). TeNT H C axonal carriers do not accu- mulate acidic tracers but rather display a neutral pH (Bohnert and Schiavo, 2005). Furthermore, the progres- sion of TeNT H C carriers along axons requires the coor- dination of both, cytoplasmic dynein, a microtubule- based motor, and myosin Va, an F actin-based motor (Hafezparast et al., 2003; Lalli et al., 2003). In spite of the above findings, several questions remain unan- swered about the nature of these axonal carriers, their biogenesis, and targeting to the soma. In this report, we demonstrate that TeNT H C enters a retrograde pathway shared by neurotrophins and their receptors. We then focus our studies on the role of two proteins emerging from our magnetic isolation ap- proach, the small GTPases Rab5 and Rab7. Rab pro- teins have been shown to play crucial functions in the regulation of intracellular transport (Zerial and McBride, 2001) and in determining cargo progression through the endosomal system (Rink et al., 2005). Here we show that Rab5 and Rab7 act in a sequential manner in controlling an axonal retrograde transport pathway in MN. In partic- ular, Rab7 is associated with a pool of retrograde car- riers in MN and dorsal root ganglia (DRG) neurons, sug- gesting that TeNT H C and neurotrophin receptors use Rab7-positive organelles to be actively transported toward the soma. Results TeNT H C Is Transported along the Neurotrophin Route TeNT H C is internalized at the NMJ and then specifically targeted to the retrograde transport route toward the MN soma, entering vesicular carriers with unknown physiological function. Previous studies had demon- strated that TeNT H C is cotransported with NGF in MN axons (Lalli and Schiavo, 2002). Here, we asked whether *Correspondence: [email protected] 5 These authors contributed equally to this work.
13

Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Apr 21, 2023

Download

Documents

Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Neuron 52, 293–305, October 19, 2006 ª2006 Elsevier Inc. DOI 10.1016/j.neuron.2006.08.018

Rab5 and Rab7 Control Endocytic Sortingalong the Axonal Retrograde Transport Pathway

Katrin Deinhardt,1,5 Sara Salinas,1,5

Carole Verastegui,1 Rose Watson,2

Daniel Worth,1 Sarah Hanrahan,3

Cecilia Bucci,4 and Giampietro Schiavo1,*1Molecular NeuroPathobiology Laboratory2Electron Microscopy Laboratory3Protein Analysis LaboratoryCancer Research UK London Research Institute44 Lincoln’s Inn FieldsLondon, WC2A 3PXUnited Kingdom4Dipartimento di Scienze e Tecnologie

Biologiche ed AmbientaliUniversita di Lecce, via Provinciale Monteroni73100 LecceItaly

Summary

Vesicular pathways coupling the neuromuscular junc-

tion with the motor neuron soma are essential for neu-ronal function and survival. To characterize the organ-

elles responsible for this long-distance crosstalk, wedeveloped a purification strategy based on a fragment

of tetanus neurotoxin (TeNT HC) conjugated to para-magnetic beads. This approach enabled us to identify,

among other factors, the small GTPase Rab7 as a func-tional marker of a specific pool of axonal retrograde

carriers, which transport neurotrophins and theirreceptors. Furthermore, Rab5 is essential for an early

step in TeNT HC sorting but is absent from axonallytransported vesicles. Our data demonstrate that

TeNT HC uses a retrograde transport pathway sharedwith p75NTR, TrkB, and BDNF, which is strictly depen-

dent on the activities of both Rab5 and Rab7. There-fore, Rab7 plays an essential role in axonal retrograde

transport by controlling a vesicular compartmentimplicated in neurotrophin traffic.

Introduction

Neurons represent the ultimate example of polarizedcells. The maintenance of their unique morphology,which is based on axonal and dendritic arborization, isnecessary for neuronal function and requires a complexnetwork of trafficking pathways to allow efficient com-munication over long distances (Goldstein and Yang,2000). Axonal transport constitutes the backbone oflong-range crosstalk in neurons and is essential for theirsurvival and differentiation. One of the most convincingdemonstrations of the crucial role of axonal transport inneuronal homeostasis is the causal link between defectsin this process and neurodegeneration (Holzbaur, 2004).In particular, key components of retrograde transportroutes, which are responsible for the targeting of neuro-

*Correspondence: [email protected] These authors contributed equally to this work.

trophins and their survival signals to the soma, havebeen associated with motor neuron disease in humansand mice (Hafezparast et al., 2003; Puls et al., 2003).Despite the importance of these axonal compartments,little is known about their nature and the machinery reg-ulating retrograde transport in neurons.

To date, few attempts to purify axonal retrogradetransport compartments have been carried out. Re-cently, Delcroix et al. have provided a preliminary char-acterization of Trk-positive signaling endosomes, whichcontain activated Erk1/2, p38, and Akt, using a sciaticnerve chamber approach (Delcroix et al., 2003). In addi-tion, several signaling proteins triggered by nerve injuryand undergoing retrograde transport have been identi-fied by a proteomic analysis in Lymnaea (Perlson et al.,2004). Here, we sought to characterize the axonal retro-grade compartment of spinal cord motor neurons (MN)by setting up a purification protocol based on paramag-netic iron beads (Fe beads) covalently bound to a frag-ment of tetanus neurotoxin (TeNT HC). TeNT HC bindswith high affinity to MN and is endocytosed by a specificclathrin-dependent pathway linked to a retrograde routeto the central nervous system (Deinhardt et al., 2006;Roux et al., 2005). TeNT HC axonal carriers do not accu-mulate acidic tracers but rather display a neutral pH(Bohnert and Schiavo, 2005). Furthermore, the progres-sion of TeNT HC carriers along axons requires the coor-dination of both, cytoplasmic dynein, a microtubule-based motor, and myosin Va, an F actin-based motor(Hafezparast et al., 2003; Lalli et al., 2003). In spite ofthe above findings, several questions remain unan-swered about the nature of these axonal carriers, theirbiogenesis, and targeting to the soma.

In this report, we demonstrate that TeNT HC entersa retrograde pathway shared by neurotrophins and theirreceptors. We then focus our studies on the role of twoproteins emerging from our magnetic isolation ap-proach, the small GTPases Rab5 and Rab7. Rab pro-teins have been shown to play crucial functions in theregulation of intracellular transport (Zerial and McBride,2001) and in determining cargo progression through theendosomal system (Rink et al., 2005). Here we show thatRab5 and Rab7 act in a sequential manner in controllingan axonal retrograde transport pathway in MN. In partic-ular, Rab7 is associated with a pool of retrograde car-riers in MN and dorsal root ganglia (DRG) neurons, sug-gesting that TeNT HC and neurotrophin receptors useRab7-positive organelles to be actively transportedtoward the soma.

Results

TeNT HC Is Transported along theNeurotrophin Route

TeNT HC is internalized at the NMJ and then specificallytargeted to the retrograde transport route toward theMN soma, entering vesicular carriers with unknownphysiological function. Previous studies had demon-strated that TeNT HC is cotransported with NGF in MNaxons (Lalli and Schiavo, 2002). Here, we asked whether

Page 2: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Neuron294

Figure 1. TeNT HC and Neurotrophin Recep-

tors Share Axonal Retrograde Carriers

MN were incubated with (A) Alexa 488-TeNT

HC and Cy3-labeled anti-p75NTR(EC) or (B)

microinjected with TrkB-GFP and then incu-

bated with Alexa 647-TeNT HC and Cy3-

labeled anti-p75NTR(EC) for 30 min and

imaged. Individual frames from a confocal

time series are shown. Cell bodies are lo-

cated out of view to the right. Arrows and ar-

rowheads point out TeNT HC carriers positive

for the neurotrophin receptors. See also

Movie S1. (C) Western blot analysis of

BDNF-GFP. BDNF-GFP-TEV-His6 (10 ng)

was purified from HEK293 conditioned me-

dium with Ni-NTA agarose beads and loaded

on a 10% SDS-PAGE together with recombi-

nant BDNF (20 ng) and an extract of HEK293

cells expressing GFP-TEV-His6 (GFP). Identi-

cal panels were probed with anti-BDNF (left)

or anti-GFP (right) antibodies. (D) The biolog-

ical activity of BDNF-GFP was assessed by

testing its ability to activate CREB in TrkB-

RFP-expressing PC12 cells. PC12 cells were

transfected with a plasmid encoding TrkB-

RFP for 24 hr and incubated for 1 hr with

partially purified BDNF-GFP from HEK293 su-

pernatant, fixed, stained for phospho-CREB

(p-CREB), and imaged. Scale bar, 10 mm. (E)

TrkB-RFP expressing MN were incubated

with partially purified BDNF-GFP for 30 min

on ice. MN were washed, warmed up, and

imaged. (F) MN were incubated with Cy3-

labeled anti-p75NTR(EC), Alexa 647-TeNT

HC, and BDNF-GFP for 30 min on ice,

washed, warmed to 37�C, and imaged. Indi-

vidual frames of a movie are shown. The cell

bodies are located out of view to the right.

Arrows point out a TeNT HC carrier positive

for the neurotrophin and p75NTR. Scale bars,

5 mm.

this compartment is a general neurotrophin transportcompartment and investigated whether it is sharedwith other neurotrophins, such as BDNF, and their re-ceptors, p75NTR and Trks. Of the Trk receptors, onlyTrkB and -C are expressed in MN at early developmentalstages (Yan et al., 1993, and data not shown), implyingthat p75NTR might be the only NGF receptor in this cellu-lar system. Therefore, we raised an antibody against theextracellular domain of p75NTR [anti-p75NTR(EC)], whichspecifically recognized p75NTR (see Figures S1B–S1D inthe Supplemental Data available online) but did not elicitreceptor transport (Figure S1E). In contrast, this p75NTR

antibody was actively transported in MN upon stimula-tion with NGF (Figure S1F) or TeNT HC (Figure S1G).The kinetic of appearance of moving organelles contain-ing p75NTR closely overlapped with that of TeNT HC-positive structures, with carriers becoming clearlyvisible after 45 min of internalization. Interestingly, MNincubated with anti-p75NTR(EC) and TeNT HC showeda high frequency of double-positive carriers (95.6%overlap, n = 118 carriers) (Figures 1A and S1H).

To assess whether Trk receptors are also sorted tothis transport compartment, we microinjected a plasmidencoding for TrkB-GFP in MN and visualized the traffick-ing of this fusion protein in relation to TeNT HC and anti-p75NTR(EC). In MN expressing TrkB-GFP, the vast ma-jority of TeNT HC carriers were positive for all threemarkers (95.5% overlap, n = 88 TeNT HC carriers) (Fig-ure 1B and Movie S1), implying that p75NTR and TrkBare transported along the MN axon within the sameorganelle, accessible to TeNT HC. This result was con-firmed by the colocalization in axonal carriers of TeNTHC and endogenous TrkB using a TrkB-specific anti-body (data not shown). The notion that p75NTR and Trkreceptors enter the same endosomal structures at latetime points (R30 min) is in agreement with a previousreport showing the accumulation of TrkA and p75NTR

in common perinuclear organelles upon neurotrophinstimulation in PC12 cells (Bronfman et al., 2003). Finally,we asked whether the physiological ligand for TrkB,BDNF, could also enter these carriers. For this purpose,we expressed a tagged version of BDNF-GFP in HEK293

Page 3: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Rab5 and Rab7 Regulate Axonal Transport295

Figure 2. Rab7 Is Associated with TeNT HC

Carriers Purified by Magnetic Isolation

(A) TeNT HC-coupled Fe beads labeled with

Alexa 488 were internalized in mouse MN

(DIV6) and imaged by low-light time-lapse

fluorescence microscopy. Inverted time se-

ries images acquired every 5 s are shown.

The cell body is visible at the bottom (see

Movie S2). Scale bar, 10 mm.

(B) Fe beads coupled with the PH domain of

cytohesin I (mock beads) or with TeNT HC

were incubated with MN (DIV6) isolated

from 25 mouse embryos. Detergent-free MN

extracts were used for the magnetic purifica-

tion, and protein bands were analyzed by

mass peptide fingerprint using a MALDI

TOF-TOF analyser (see Table S1; NI, non-

identified band).

(C) Similar samples obtained from MN cul-

tures derived from 16 rat spinal cords were

subjected to Western blot and probed with

the antibodies indicated. The input lanes cor-

respond to 1% of postnuclear supernatant of

each sample blotted for Rab5.

(D and E) After internalization of TeNT HC for

60 min at 37�C, MN were fixed and immuno-

stained for endogenous Rab5 (D) or Rab7

(E). TeNT HC and Rab5 or TeNT HC and

Rab7 colocalized in axonal puncta (arrows).

Scale bar, 5 mm.

cells. This fusion protein, which was recognized by bothanti-BDNF and anti-GFP antibodies (Figure 1C), is bio-logically active, since it was able to induce CREB phos-phorylation in PC12 cells (Figure 1D). Moreover, bindingof BDNF-GFP to MN was enhanced by overexpressionof TrkB (Figure 1E), whereas it was negligible in copuri-fied nonneuronal cells (data not shown), suggesting thatthe interaction of BDNF-GFP with the MN surface wasspecific. Upon incubation of MN with BDNF-GFP, anti-p75NTR(EC), and TeNT HC, we found that most of theTeNT HC carriers were positive for anti-p75NTR(EC) andBDNF-GFP (90.1% overlap, n = 71 TeNT HC carriers)(Figure 1F). Altogether, these observations strongly sug-gest that TeNT HC is retrogradely transported along theneurotrophin/neurotrophin receptor transport route inMN axons.

Isolation of Axonal Retrograde Carriers using TeNTHC-Conjugated Magnetic Beads

To further characterize these axonal carriers, we chosea purification protocol based on TeNT HC coupled tomagnetic nanobeads (Fe beads). First, we verified thatimmobilized TeNT HC conserved the binding and trans-

port properties of the unconjugated protein. For this, weprepared a traceable version of the magnetic particlesby using Alexa 488-TeNT HC. TeNT HC-coupled Febeads bound specifically to discrete puncta on the sur-face of MN (Figures S2A–S2D). Exploiting the high elec-tron density of these beads, we were able to monitortheir entry in vesicular or tubular structures along theaxon by EM (Figures S2G–S2J). Larger organelles con-taining internal membranes were also labeled, althoughat lower frequency (Figures S2E and S2F). In addition,TeNT HC Fe beads were transported in a retrogradefashion along the axon, as shown by time-lapse micros-copy (Figure 2A and Movie S2), indicating that TeNTHC-coupled Fe beads are targeted to tubulo-vesicularorganelles in spinal cord MN, as previously reportedfor unconjugated TeNT HC (Lalli et al., 2003). In contrast,Fe beads bound to an unrelated protein, the cysteine-tagged pleckstrin-homology (PH) domain of cytohesinI (mock Fe beads), did not bind to MN nor underwentretrograde transport under these conditions (data notshown).

For a proteomic characterization of these axonal car-riers, spinal cord MN (5–7 days in vitro [DIV5–7]) were

Page 4: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Neuron296

incubated with TeNT HC or mock Fe beads, mechani-cally disrupted in detergent-free medium, and the result-ing extracts passed through a high magnetic field de-vice. The proteins recovered with either TeNT HC ormock Fe beads were then analyzed by SDS-PAGE.Comparison of the silver-stained gels allowed the iden-tification of 17 major specific bands (Figure 2B), whichgave 16 unique protein sequences by MALDI TOF-TOF. The experiment was repeated with TeNT HC Febeads prepared independently, and 9 proteins out of16 identified sequences were identical in both experi-ments. The proteins identified by this approach belongto three functional classes: signaling (2), vesicular trans-port/cytoskeleton (7), and chaperones/translocation (5)(Table S1). Although most of the proteins associatedwith the TeNT HC carriers are likely to play a role in axo-nal transport based on their known function in other sys-tems, for the scope of this work we focused on the smallGTPase Rab7, due to its pleiotropic roles in regulatingthe endosomal pathway and the emerging link with pa-thologies of the nervous system (Houlden et al., 2004;Verhoeven et al., 2003). The specific recruitment ofRab7 to TeNT HC carriers was verified by Western blotanalysis of the purified beads (Figure 2C). As predicted,both p75NTR and endogenous TrkB were associatedwith TeNT HC carriers, confirming the notion that TeNTHC enters the neurotrophin receptor transport route(Figure 1). Erk1/2 and phospho-Erk1/2 were also de-tected on TeNT HC beads, suggesting that these endo-somes may have signaling capability (Figure 2C). In ad-dition, we found that TeNT HC beads bound Rab5,a small GTPase of the endocytic pathway, which actsupstream of Rab7 (Zerial and McBride, 2001). The inter-action of Rab5 and Rab7 with TeNT HC-containingorganelles was confirmed by immunofluorescence,showing that TeNT HC-positive structures partially colo-calized with endogenous Rab5 (29.6% of the total;Figure 2D) and Rab7 (28.2%; Figure 2E) in MN axons.This level of colocalization was well above that observedfor Rab3, a Rab protein involved in a distinct exo-endo-cytic pathway in neurons (7.0%; Table S2), suggestingthat Rab5 and Rab7 specifically bound to a pool ofTeNT HC-containing organelles.

Rab5 Regulates an Early Sorting Step Preceding

Axonal TransportRab GTPases and their effectors are primary determi-nants of compartmental specificity in eukaryotic cells.In the endocytic pathway, Rab5 and Rab7 act in asequential manner (Rink et al., 2005; Vonderheit andHelenius, 2005), with Rab5 controlling early step(s),including clathrin-dependent endocytosis, targeting ofcargoes to early endosomes, and endosome fusion.Rab7 instead acts at later stages, regulating cargo pro-gression from early to late endosomes (Pfeffer, 2003;Zerial and McBride, 2001). Interestingly, Rab5 hasbeen suggested to be associated with neurotrophin sig-naling endosomes (Delcroix et al., 2003). Therefore, wedecided to first explore a potential role of Rab5 inTeNT HC transport.

Live imaging of GFP-Rab5wt-expressing MN revealedthat Rab5wt concentrated in bright puncta along theaxon. These structures were stationary or displayed lo-calized short-range movement (Figure 3A and Movie

S3). As an alternative representation of the time-lapseanalysis, a kymograph was generated through drawinga line along the axon and stacking pixels derived fromsubsequent frames of the video as a function of time(Figure 3B). Stationary compartments appear as a verti-cal line in the resulting diagram (e.g., dotted lines inFigure 3B), while moving carriers are represented by di-agonal traces (e.g., dotted lines in Figure 4B). The angleof these traces is proportional to the speed and directionof the moving organelle. The representative kymographin Figure 3B showed that no GFP-Rab5wt-positive com-partments underwent long-range transport in MNaxons. Furthermore, analysis of TeNT HC transport inGFP-Rab5wt-expressing neurons showed that many os-cillatory TeNT HC vesicles were positive for GFP-Rab5wt

(Figure 3C, asterisks), whereas carriers undergoinglong-range retrograde axonal transport did not containGFP-Rab5wt (Figure 3C, arrow and arrowhead, andMovie S4).

To further investigate whether Rab5 activity playsa functional role in the sorting of TeNT HC to the retro-grade transport route, we expressed the constitutive-active (GFP-Rab5Q79L) and dominant-negative (GFP-Rab5N133I) forms of Rab5 in MN. Overexpression ofeither wild-type Rab5 or its Rab5Q79L mutant did notalter the transport-speed profile of TeNT HC vesicles(Figure S3A) and had no obvious effect on the frequencyof transported carriers. The GFP-Rab5Q79L mutant isfully functional in MN, since it induced the appearanceof enlarged endocytic structures in the soma and den-drites (Figure S4A), as previously shown in nonneuronalcells (Stenmark et al., 1994). However, no swollen endo-somes could be found in axons (Figure S4B). This differ-ence may be due to the absence of the Rab5 effectorEEA1 from MN axons (Figure S4C, arrow), a finding re-ported in hippocampal neurons (Wilson et al., 2000). Incontrast to GFP-Rab5Q79L, which did not affect TeNTHC trafficking, overexpression of the dominant-negativemutant GFP-Rab5N133I in MN completely abolished ret-rograde transport of TeNT HC (Figure 3E). To monitorcell viability at the time of imaging, MN were stainedwith tetramethylrhodamine ethyl ester (TMRE), a dyethat accumulates in mitochondria with an intact mem-brane potential. As shown in Figure 3F, TMRE uptakeand mitochondria movement were unaffected, confirm-ing the specificity of the inhibition. Since Rab5 has beeninvolved in early steps of endocytosis (Bucci et al., 1992;McLauchlan et al., 1998), we tested whether the TeNT HC

pathway was already impaired at the stage of cell entryin MN expressing GFP-Rab5N133I. MN were incubatedwith a disulfide-linked biotin-TeNT HC conjugate andthen treated with a membrane-impermeable reducingagent in order to eliminate biotin from the surface-boundtoxin (Deinhardt et al., 2006). As shown in Figure 3D,biotin staining persisted in GFP-Rab5N133I-expressingMN, indicating that Rab5 activity is not essential forTeNT HC uptake but is required for a subsequent steppreceding the onset of fast axonal retrograde transport.

Rab7 Regulates Long-Range Retrograde Axonal

Transport in MNNext, we investigated the involvement of Rab7 in TeNTHC transport. Similarly to GFP-Rab5wt, GFP-Rab7wt con-centrated in bright puncta along MN axons (Figure 4A).

Page 5: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Rab5 and Rab7 Regulate Axonal Transport297

Figure 3. Rab5 Displays Local Movement in

MN Axons and Regulates an Early Event in

TeNT HC Trafficking

GFP-Rab5wt displays a punctuate pattern

along axons (A) and is mainly found in station-

ary vesicles (A and B). Shown are individual

frames from a confocal time series of GFP-

Rab5wt in a MN axon (A) and the correspond-

ing kymograph (B). Dotted lines indicate two

examples of oscillating GFP-Rabwt puncta.

The MN soma is out of view to the right. See

also Movie S3. (C) GFP-Rab5wt and TeNT HC

colocalize on stationary structures in MN

axons. Individual frames from a confocal

time series displaying dual-positive GFP-

Rab5wt and Alexa 555-TeNT HC still compart-

ments (asterisks) and Alexa 555-TeNT HC

moving carriers (arrows and arrowheads)

are shown. The MN soma is out of view to

the right. See also Movie S4. (D) TeNT HC in-

ternalization is not blocked by GFP-Rab5N133I

overexpression. MN were microinjected with

a plasmid encoding GFP-Rab5N133I, and after

20 hr of expression incubated with biotiny-

lated TeNT HC, MESNA-treated, fixed, and

stained for remaining biotin. (E and F) Rab5

activity is required for TeNT HC transport.

MN expressing GFP-Rab5N133I were incu-

bated with Alexa 647-TeNT HC (E) and

TMRE (F) and imaged. Shown are the kymo-

graphs of the individual channels. Selected

optical sections of axons are shown on top

of the kymographs. The soma is located out

of view to the right. Scale bars, 5 mm; in (D),

20 mm.

However, in contrast to GFP-Rab5wt-positive organ-elles, the majority of GFP-Rab7wt puncta displayedlong-range bidirectional movement, which was stronglyretrogradely biased (Figure 4B and Movies S5 and S6).Many TeNT HC carriers (43% of the total) were also pos-itive for GFP-Rab7wt (Figure 4C and Movie S6), and eventhough GFP-Rab7wt showed bidirectional movement inthe axon, this subset of double-positive organelleswas exclusively moving in the retrograde direction. Ex-pression of GFP-Rab7wt did not alter TeNT HC transport,since the speed distribution of TeNT HC carriers in MNexpressing GFP-Rab7wt was similar to that seen in con-trol cells (Figure S3B).

Kinetic analysis showed that GFP-Rab7wt/TeNT HC

carriers were characterized by a relatively slow speedranging between 0.2 and 1.2 mm/s (Figure 4D, graybars), which matched the overall speed profile of GFP-Rab7wt vesicles (data not shown). The speed distribu-tion of the GFP-Rab7wt/TeNT HC organelles and theentire pool of TeNT HC vesicles (Figure 4D) were furtheranalyzed by applying a multiple Gaussian curve fit. Asreported previously (Hafezparast et al., 2003; Lalliet al., 2003), the speed profile of these carriers is bestdescribed by the sum of three components centeredat 0 mm/s (stationary/pausing), 0.53 mm/s (intermediate),and 1.15 mm/s (fast). Analysis of the contribution of thesespeed components to the overall retrograde transportrevealed that the entire intermediate component ofTeNT HC carriers was positive for GFP-Rab7wt, whereas

only 40% of the fast component was double positive(Figure S3C, gray bars). These results demonstratedthat Rab7 is targeted to a discrete population of axonalretrograde carriers characterized by distinct kineticproperties.

To investigate the functional significance of Rab7 onTeNT HC carriers, we expressed the constitutive-active (GFP-Rab7Q67L) and dominant-negative (GFP-Rab7N125I) mutants in MN. As for Rab5, expression ofthe wild-type or constitutive-active Rab7 had no obvi-ous effects on organelle trafficking. TeNT HC partiallycodistributed with GFP-Rab7Q67L both in cell bodiesand axons (Figures S4D and S4E), and TeNT HC axonalcarriers displayed a speed profile overlapping that ofcontrol cells (Figure S3B). In contrast, the axonal trans-port of TeNT HC was blocked upon GFP-Rab7N125I

expression (Figure 4E and Movie S7). As shown in thekymograph of Figure 4E, TeNT HC carriers were immo-bile or displayed short-range oscillatory movements,suggesting that, despite the association of Rab7wt witha discrete subset of TeNT HC transport compartments,Rab7 activity was required for the retrograde transportof the whole population of TeNT HC carriers. This blockwas specific, since mitochondrial morphology and dy-namics were unaffected in GFP-Rab7N125I expressingMN (Figure 4F and Movie S8).

The localization of Rab5 to oscillatory organelles andRab7 to long-range carriers suggested that Rab5 actsupstream of Rab7 in the endocytic sorting of TeNT HC,

Page 6: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Neuron298

Figure 4. GFP-Rab7wt and TeNT HC Are

Cotransported in Retrograde Carriers

(A and B) GFP-Rab7wt is found in discrete

puncta along the axon and is transported

over long distances in MN. Individual frames

from a confocal time series (A) and the corre-

sponding kymograph (B) show GFP-Rab7wt-

positive carriers in a MN axon. Dotted lines

in (B) indicate two examples of transported

GFP-Rab7wt puncta. The MN soma is out of

view to the right. See also Movie S5.

(C) GFP-Rab7wt and TeNT HC are cotrans-

ported in retrograde carriers. Confocal time

series frames show a dual-positive GFP-

Rab7wt and Alexa 555-TeNT HC carrier mov-

ing along a MN axon (arrows). The MN soma

is out of view to the right. See also Movie S6.

(D) GFP-Rab7wt/TeNT HC carriers represent

a discrete pool of retrograde transport organ-

elles, as shown by comparing the speed pro-

file of the total population of TeNT HC carri-

ers (black circles; 1039 single movements,

126 carriers) with the double-positive GFP-

Rab7wt/TeNT HC organelles (gray bars; 418

single movements, 52 carriers). Speed pro-

files were assembled by binning the speed

values using a 0.2 mm/s window and cal-

culating the relative frequencies. Retrograde

transport is conventionally shown as positive,

and pauses during movement are grouped

at 0 mm/s.

(E and F) MN were injected with the domi-

nant-negative GFP-Rab7N125I mutant (in

green) for 18 hr and then incubated with Alexa

647-TeNT HC (E) and TMRE (F) for 30 min at

37�C. Under these conditions, TeNT HC

puncta were oscillatory or stationary, as

shown in the kymograph (E) (see Movie S7),

while mitochondrial motility was not affected

(F) (see Movie S8). Selected optical sections

of axons are shown on top of the kymo-

graphs. The soma is out of view to the right.

Scale bars, 5 mm.

as has been established for other ligands in different celltypes (Chavrier et al., 1990; Lakadamyali et al., 2006;Rink et al., 2005; Vonderheit and Helenius, 2005). To ver-ify this hypothesis, we incubated MN with TeNT HC for15 min, a time point before the onset of retrograde trans-port, or 60 min, at which time retrograde transport is wellunder way. Cells were then fixed and stained for endog-enous Rab5 or Rab7. Quantification of double-positivestructures showed that after 15 min of internalization33.4% of the TeNT HC vesicles labeled for Rab5 (n =602) while only 15.5% were also positive for Rab7 (n =568). However, after 60 min of continuous incubationwith the toxin, the percentage of TeNT HC organellescontaining Rab7 increased to 28% (n = 719), whereasthe proportion of Rab5/TeNT HC structures remainedunchanged (29.6%; n = 601) (Table S2).

Taken together, these results indicate that Rab5 andRab7 act in a sequential manner in the biogenesis andprogression of axonal retrograde carriers. Rab5 is im-portant for the early sorting of TeNT HC, probably at astep following internalization but preceding transport,whereas Rab7 controls a later event, ensuring the move-ment of these organelles or determining the onset oftransport itself. Despite its localization to a discretepool of TeNT HC carriers, Rab7 is indeed required for

the integrity of TeNT HC retrograde transport as a whole,suggesting its involvement in a primary regulatory eventof this pathway.

Functional Rab7 Is Required for NeurotrophinReceptor Transport

Since we had shown above that TeNT HC shares a trans-port compartment with both p75NTR and TrkB (Figure 1),we tested whether neurotrophin receptor transport wasalso regulated by Rab7. First, we looked at the transportof endogenous p75NTR or TrkB-RFP in GFP-Rab7wt-expressing MN. To this end, MN were injected witha GFP-Rab7wt plasmid and, upon expression, incubatedwith Cy3-labeled anti-p75NTR(EC). Axonal transport ofthe labeled antibody was triggered by addition of NGF.Alternatively, MN were injected with GFP-Rab7wt andTrkB-RFP, incubated with TeNT HC, and imaged. Asexpected, GFP-Rab7wt was detected on a subset ofp75NTR-positive moving structures (31% of p75NTR-containing carriers) (Figure 5A and Movie S9) and ona subpopulation of retrograde TrkB-RFP carriers (28%;Figure 5B and Movie S10). In a similar way to TeNT HC,expression of GFP-Rab7N125I led to the almost completeblockade of the transport of both p75NTR and TrkB-RFP(Figures 5C and 5D).

Page 7: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Rab5 and Rab7 Regulate Axonal Transport299

Figure 5. Rab7 Regulates the Long-Range

Axonal Retrograde Transport of Neurotro-

phin Receptors

MN were injected with GFP-Rab7wt (in green)

(A) or GFP-Rab7N125I (C) or GFP-Rab7wt and

TrkB-RFP (in red) (B) or GFP-Rab7N125I and

TrkB-RFP (D). Samples were incubated with

Cy3-labeled anti-p75NTR(EC) (in red) and

100 ng/ml NGF (A and C) or 40 nM TeNT HC

(B and D) and imaged. Frames were acquired

every 5 s. Shown are kymographs of Movies

S9 and S10. Cell bodies are out of view to

the right. Arrowheads show the beginning

and end of the trace of a single-positive

p75NTR carrier (A) or a TrkB carrier (B). Aster-

isks indicate the trace of double-positive

organelles. Scale bars, 5 mm.

The observation that inhibiting Rab7 activity blocksthe retrograde transport of neurotrophin receptors ledto the question whether expression of Rab7N125I also in-terferes with known nuclear responses to neurotrophinsignaling. To test this hypothesis, we expressed GFP-Rab7wt or GFP-Rab7N125I in MN, mass treated themwith BDNF, and then monitored activation of the tran-scription factor CREB. While serum- and neurotrophin-starved cells showed minimal CREB phosphorylation(Figure 6A, top panel, asterisks), exposure to BDNF trig-gered CREB activation in MN (Figure 6A, middle andbottom panels, asterisks). Expression of either GFP-Rab7wt or GFP-Rab7N125I did not alter the activationof CREB by BDNF (Figure 6A, bottom panel, arrow),as confirmed by quantification of phospho-CREB (p-CREB) in control, GFP-Rab7wt, or GFP-Rab7N125I ex-pressing MN (Figure 6B). This observation is in agreementwith previous results obtained for NGF signaling in PC12cells (Saxena et al., 2005a) and suggests that interferingwith Rab7 function in MN is not sufficient to block thenuclear response to mass neurotrophin stimulation.

Rabs and Axonal Retrograde Transport

in Sensory NeuronsOne question emerging from these findings is whetherthe axonal transport features described above are spe-cific for MN or are conserved in other neuronal types. Aprevious study suggested that in DRG, axonal endo-somes containing NGF were positive for Rab5 (Delcroixet al., 2003). Since TeNT HC is also taken up by sensoryneurons in vivo (Meckler et al., 1990; Stockel et al., 1975),we used this probe to examine long-range axonal trans-port in adult DRG. TeNT HC bound preferentially to largeand medium diameter neurons (soma R 10 mm), which

have been reported to express p75NTR and Trk receptors(Tucker et al., 2005). Accordingly, of the DRG that bindTeNT HC, 85% were positive for p75NTR, 87% for TrkB,and 66% for TrkA (data not shown).

First, we imaged transport of TeNT HC together withanti-p75NTR(EC). Just like in MN, both probes weretransported exclusively in the retrograde direction andlargely colocalized (83% overlap, n = 91 TeNT HC car-riers; Figure 7A), suggesting that TeNT HC enters theneurotrophin receptor transport route also in DRG. Wethen assessed the dynamics of Rab5 and Rab7 in theseneurons. Similarly to the pattern observed in MN, GFP-Rab5wt displayed a punctate distribution along theaxon (Figure 7B), and live imaging revealed that thesecompartments were stationary or underwent short-range movements (Figures 7B and 7C and Movie S11).Some of the stationary TeNT HC structures were positivefor GFP-Rab5wt (data not shown), but carriers did notcontain this marker (Figure 7B, arrows). In contrast,GFP-Rab7wt-containing organelles, some of whichwere positive for TeNT HC, displayed long-range axonaltransport, which was strongly retrogradely biased (Fig-ure 7D, arrows, Figure 7E, and Movie S12). The dynam-ics of GFP-Rab5 and GFP-Rab7 organelles were similarin TeNT HC-negative DRG (data not shown).

These results demonstrate that TeNT HC enters an en-docytic pathway used by components of the neurotro-phin signaling cascade to undergo axonal retrogradetransport in MN and DRG. This pathway requires theconcerted activities of Rab5 and Rab7 during sortingsteps prior to, or at the onset of, the movement ofaxonal carriers, suggesting a role for Rab7 as a generalregulator of endocytic long-range axonal retrogradetransport.

Page 8: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Neuron300

Discussion

Rab5 and Rab7 Regulate a Fast AxonalRetrograde Pathway

The use of bacterial protein toxins has allowed the mo-lecular characterization of several cellular pathways(Schiavo and van der Goot, 2001). In this study, we dem-onstrate that the binding fragment of TeNT enters a ret-rograde transport route used by neurotrophins as wellas TrkB and p75NTR, thus suggesting that this probe isa functional marker for the axonal trafficking of neurotro-phins and their receptors. The nature and regulation ofthese transport pathways are of crucial interest sincetheir impairment has been linked to neurodegeneration(Guzik and Goldstein, 2004; Holzbaur, 2004). Using para-

Figure 6. Expression of GFP-Rab7N125I Does Not Impair BDNF-

Dependent CREB Activation

MN were microinjected with plasmids encoding GFP-Rab7wt or

GFP-Rab7N125I. After 12–14 hr of expression, cells were starved for

5 hr and then stimulated with 2 ng/ml BDNF. Cells were stained

for p-CREB, fixed, and imaged. (A) Starved MN nuclei do not label

for p-CREB (top panel, asterisks). Treatment with BDNF activates

CREB in control cells (middle and bottom panels, asterisks) as

well as in GFP-Rab7wt- (middle panel, arrows) and GFP-Rab7N125I-

expressing MN (bottom panel, arrows). Scale bar, 50 mm. (B) Quan-

tification of CREB activation expressed as a box and whiskers

diagram (a versus b, c, and d, p % 0.002; b versus c and d, p R

0.05). Numbers in brackets indicate the MN observed per condition.

magnetic beads linked to TeNT HC, we have purified in-tact retrograde carriers from MN and identified Rab7 asan essential component of these organelles. We showthat the sequential activities of Rab5 and Rab7 are re-quired for coupling specialized clathrin-dependent en-docytosis to fast retrograde axonal transport (Figure 8).In particular, we observe Rab5 associated with station-ary or oscillatory organelles, whereas Rab7 localizes toa subpopulation of moving compartments. Despite thislocalization to a specific pool of TeNT HC carriers, im-pairment of Rab7 function leads to a complete blockadeof TeNT HC transport, implying a tight control by Rab7on a step that precedes or coincides with the recruit-ment of the carriers onto cytoskeletal tracks.

Conversion from Rab5 to Rab7 is emerging as an im-portant mechanism of cargo progression from early tolate endosomes. Two models have been recently pro-posed whereby Rab7 is targeted to endosomal struc-tures during endocytic maturation either in presence ofRab5 (Vonderheit and Helenius, 2005) or when Rab5 dis-sociates from these structures (Rink et al., 2005). Thisprogression from Rab5- to Rab7-positive endosomesis also critical for the entry of various infectious agents,such as HIV (Vidricaire and Tremblay, 2005) and Semlikivirus (Vonderheit and Helenius, 2005). Our observationsthat Rab5-positive puncta are mostly stationary,whereas Rab7 is present in moving organelles, suggestthat Rab5 acts upstream of Rab7 and that the conversionfrom Rab5- to Rab7-positive structures could control thegeneration of axonal retrograde carriers (Figure 8). Thismodel involving a Rab cascade in the regulation axonaltransport is further supported by the fact that the recruit-ment of Rab7 on TeNT HC-containing organelles in-creases over time after internalization (Table S2).

Recent evidence indicates that two populations ofRab5-positive early endosomes exist: a smaller groupthat is highly dynamic and matures rapidly by acquiringRab7 and a larger one displaying low mobility, whichmatures slowly (Lakadamyali et al., 2006). This latterpopulation resembles the GFP-Rab5wt-positive struc-tures observed in MN, which show very localized move-ment and do not shed GFP-Rab5wt during the time ofimaging. The slow maturation of TeNT HC-containingstationary endosomes is in agreement with the ob-served 45 min delay between uptake and onset of trans-port (Figure 8) and with the lag phase reported for theappearance of 125I-NGF in the soma of sympathetic neu-rons (Claude et al., 1982; Ure and Campenot, 1997). Incontrast, the highly dynamic population, which rapidlyacquires Rab7, preferentially contains cargo targetedfor degradation (Lakadamyali et al., 2006). This is inagreement with the fact that Rab7-positive organelleshave been described so far as acidic (Figure 4F) (Gruen-berg and Maxfield, 1995). In contrast, TeNT HC carriersdisplay a neutral pH and, similar to NGF-containing axo-nal organelles (Ure and Campenot, 1997) and p75NTR-positive endosomes in PC12 cells (Bronfman et al.,2003), follow a nondegradative route in MN axons (Boh-nert and Schiavo, 2005; Lalli and Schiavo, 2002). Thisnew role for Rab7 in axonal retrograde transport addsto the diverse cellular functions attributed to this smallGTPase, from the regulation of the trafficking betweenlate endosomes and lysosomes to autophagosomematuration, growth factor-dependent cell nutrition, and

Page 9: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Rab5 and Rab7 Regulate Axonal Transport301

Figure 7. Rab Dynamics and Axonal Retro-

grade Transport in DRG

(A) TeNT HC and p75NTR are cotransported in

DRG. Cells were incubated with Alexa 647-

TeNT HC (in green) and Cy3-labeled anti-

p75NTR(EC) (in red) and imaged. Frames

were taken every 5 s.

(B and C) DRG were injected with GFP-

Rab5wt, incubated with Alexa 647-TeNT HC,

and imaged by time-lapse microscopy.

Shown are four consecutive frames, taken

every 5 s (B). Arrows indicate a TeNT HC car-

rier, asterisks a stationary GFP-Rab5wt com-

partment. See also Movie S11.

(C) Kymograph of the green channel of Movie

S11. All GFP-Rab5wt compartments show re-

stricted local movement.

(D and E) DRG were injected with GFP-

Rab7wt, incubated with Alexa 647-TeNT HC,

and imaged. (D) Shown are four consecutive

frames taken every 5 s. Arrows indicate

a dual-positive TeNT HC and GFP-Rab7wt

carrier. See also Movie S12. (E) Kymograph

of the green channel of Movie S12. GFP-

Rab7wt organelles show retrogradely biased

long-range axonal transport. MN soma are

out of view to the right. Scale bars, 5 mm.

apoptosis (Bucci et al., 2000; Gutierrez et al., 2004; Har-rison et al., 2003; Jager et al., 2004; Snider, 2003).

Rabs and the Signaling EndosomeA previous report suggested a role for Rab5 and its ef-fector EEA1 in the retrograde transport and signaling

of NGF in embryonic DRG and sciatic nerve (Delcroixet al., 2003). In MN, we found EEA1 in the soma and den-drites, but not in axons, consistent with previous reportson EEA1 distribution in hippocampal neurons (Wilsonet al., 2000). This restricted localization might explainthe absence of enlarged endosomes from axons of

Figure 8. A Specific Endocytic Pathway

Coupled to the Retrograde Transport Route

in MN

Upon internalization via clathrin-mediated

endocytosis, TeNT HC is sorted toward the

retrograde transport route. It first transits

through a Rab5-positive, stationary compart-

ment and subsequently progresses to a

Rab7-positive moving compartment. This

process is slow, since moving carriers be-

come evident only after about 45 min after

the addition of TeNT HC. The Rab7-positive

sorting vesicles generate both Rab7-positive

intermediate speed carriers and Rab7-nega-

tive fast carriers. The transport pathway of

TeNT HC is shared by the neurotrophin recep-

tors p75NTR and TrkB, as well as their ligands

NGF and BDNF, although it is presently

unclear at which stage these pathways

converge.

Page 10: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Neuron302

GFP-Rab5Q79L expressing MN but makes it unlikely thatEEA1 plays a role in axonal transport. As proposed byDelcroix et al. (2003) for the propagation of the NGF-de-pendent signal, our study suggests that functional Rab5is indeed essential for the progression of TeNT HC retro-grade compartments along the endocytic pathway.However, since Rab5-positive puncta were found to un-dergo localized axonal movement in MN and DRG, weconcluded that Rab5 activity is required during a steppreceding long-range axonal transport.

In contrast, Rab7, which was detected in MN and DRGaxons, localizes to a subpool of TeNT HC carriers as wellas p75NTR and TrkB compartments. Impairment of itsactivity inhibits specifically the movement of TeNT HC,p75NTR, and TrkB-containing carriers, which also recruitp-Erk1/2. Based on these observations, Rab7 might actas a central regulator of neurotrophin transport and/orsignaling in neurons. Rab7-positive structures indeedcontain numerous players of the neurotrophin signalingpathway, such as TrkA (Saxena et al., 2005b) and atypi-cal PKC (Samuels et al., 2001). In PC12 cells, Rab7 im-pairment has been shown to induce the endosomal ac-cumulation of TrkA. Surprisingly, this did not lead toa decrease of NGF signaling, but to its enhancement,with an increase in TrkA and Erk1/2 phosphorylationand stimulation of neurite outgrowth in PC12 cells (Sax-ena et al., 2005a). Similarly, Rab7 inhibition in MN, withthe consequent blockade of the retrograde transportof neurotrophins, did not alter the nuclear activation ofCREB upon global exposure to BDNF. However, in ourexperimental system we cannot distinguish whetherCREB activation was triggered by distal signals propa-gated in spite of the blockade of retrograde transportor generated within the somatodendritic compartment.A previous study reported that NGF internalization wasnot required for CREB-mediated survival in PC12 cells(Zhang et al., 2000). Since it is unclear whether a similarmechanism exists for BDNF signaling, we cannot ex-clude that the activation of CREB by BDNF might be trig-gered directly from the MN cell surface.

Previous studies showed that p75NTR is proteolyticallyprocessed upon activation and one of the generatedfragments has signaling capability (Grob et al., 1985;Kanning et al., 2003), raising concerns on the integrityof the transported p75NTR in MN axons. However, wefound that full-length p75NTR was associated withTeNT HC vesicles, suggesting that this receptor is pres-ent as an intact protein on these axonal organelles andthat at least a portion is transported toward the somawithout undergoing proteolytic cleavage (Johnsonet al., 1987). Given the high number of interacting part-ners, these contrasting observations suggest thatp75NTR may undergo a different processing and triggerdifferent signaling cascades depending on the activat-ing stimulus.

Axonal Transport and Neurodegenerative DiseasesThe integrity of the axonal transport machinery is essen-tial for neuronal survival. Mutations affecting motor pro-teins or factors regulating microtubule stability havebeen linked to MN degeneration (Guzik and Goldstein,2004; Holzbaur, 2004). In light of the role of Rab5 andRab7 in the regulation of axonal transport, a predictionemerging from our studies is that an impairment of their

activity may lead to neuronal defects. This is indeed thecase, since an early-onset form of amyotrophic lateralsclerosis (ALS) is due to loss-of-function mutations inthe ALS2 gene, coding for a Rab5-specific guanine-nucleotide exchange factor termed alsin (Devon et al.,2006; Hadano et al., 2006). Moreover, mutations inRab7 have been shown to cause the motor and sensoryneuropathy type IIB, also known as Charcot-Marie-Tooth type 2B (CMT2B) (Houlden et al., 2004; Verhoevenet al., 2003).

Our findings demonstrating a crucial role for Rab5 andRab7 in endosomal sorting and axonal transport pavethe way for the study of the pathogenesis of neurode-generative diseases such as ALS and CMT2B at the mo-lecular level. Characterization of other factors identifiedusing our magnetic isolation strategy, combined withretrograde transport assays in MN, will allow us to un-cover more players in the network of interactions linkingaxonal transport, the endocytic machinery and neuronalsurvival. In this regard, it is worth noting that a complexcontaining Hsp70 and Hsp90, both found in our proteo-mic analysis, have been shown to regulate Rab-GDI ac-tivity (Sakisaka et al., 2002) and, when overexpressed,ameliorate the symptoms of motor neuron disease(Adachi et al., 2003; Kieran et al., 2004).

Experimental Procedures

Materials

BDNF-GFP tagged at the carboxyl terminus with a cleavable His6 se-

quence was prepared using as template a plasmid kindly provided

by V. Lessmann (Johannes Gutenberg University, Mainz) and ex-

pressed in HEK293 cells (see the Supplemental Experimental Proce-

dures). The TrkB-GFP and TrkB-RFP encoding plasmids were a kind

gift from M.V. Chao (Skirball Institute, New York University).

Primary antibodies were used as follows: anti-Rab5 (Synaptic

Systems), 1:150 for IF, 1:1000 for WB; anti-Rab7 (see Figure S5

and Supplemental Experimental Procedures), 1:500 for IF, 1:1000

for WB; anti-p75NTR(EC) (see Figures S1A and S1B and Supplemen-

tal Experimental Procedures), 1:5000 for IF; Cy-3 conjugated anti-

p75NTR(EC), 1:500; anti-p75NTR(IC), 1:500 for WB (see Supplemental

Experimental Procedures); anti-TrkB (BD Transduction Laborato-

ries), 1:1000; anti-Erk1/2 and anti-pErk1/2 (Cell Signaling), 1:1000;

anti-pCREB (Cell Signaling), 1:500 overnight; anti-GFP (clone 3E1,

Cancer Research UK Monoclonal Facility), 1:1000; anti-BDNF (Santa

Cruz) 1:200.

Magnetic Isolation of the Axonal Retrograde Carriers

1.25 mg of colloidal paramagnetic iron beads coated with amino-

dextran (Fe beads) were incubated with 20 mM sulfo-EMCS for

1 hr at 22�C in a final volume of 1.2 ml of PBS. Fe beads were loaded

on a 2 M sucrose cushion and spun at 265,000 3 g for 17 min at 4�C.

This step was repeated, and the final pellet was resuspended in

0.5 ml PBS. 0.9 nmoles of recombinant cysteine-tagged TeNT HC

(Lalli and Schiavo, 2002) or the PH domain of cytohesin I (mock

beads) were mixed with the activated Fe beads in the presence of

22-fold molar excess of tris[2-carboxyethylphosphine]hydrochlor-

ide in 1.25 ml of PBS and incubated for 16 hr at 4�C. The reaction

was stopped by addition of 4 mM reduced glutathione for 20 min

on ice. TeNT HC-coupled Fe beads were washed in PBS, loaded

on a sucrose cushion, and spun twice as described above. Beads

were finally resuspended in 0.5 ml of HBSS and dialyzed overnight

against HBSS. In selected experiments, TeNT HC was labeled with

Alexa 488 prior to conjugation to Fe beads. Conditions were opti-

mized to incorporate 1 mole of dye per mole of TeNT HC, thus pre-

serving three of the four cysteines of the tag for the reaction with

activated Fe beads.

Preparative MN cultures were isolated from 20 to 25 mouse em-

bryos and plated onto 100 mm dishes. Prior to binding, mock or

TeNT HC-coupled Fe beads (1.25 mg of beads/dish) were diluted

Page 11: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Rab5 and Rab7 Regulate Axonal Transport303

in cold HBSS, 0.4% BSA final, incubated for 10 min with 15 mg/ml

polyornithine, and then centrifuged at 6200 3 g for 5 min at 4�C to

remove aggregates. Precooled MN were incubated with Fe beads

for 1 hr on ice. After two washes in HBSS, 0.4% BSA, cells were in-

cubated at 37�C in complete medium for 1 hr, cooled on ice, scraped

in 1 ml HBSS supplemented with a protease inhibitor cocktail

(Roche), and centrifuged at 170 3 g for 5 min at 4�C. After resuspen-

sion in breaking buffer (BB: 0.25 M sucrose, 10 mM HEPES-KOH [pH

7.2], 1 mM EDTA, 1 mM MgOAc, complete protease inhibitors,

Roche), MN were passed 15 times through a cell cracker (18 mm

clearance; EMBL) and centrifuged at 690 3 g for 10 min at 4�C. AS

columns (Miltenyi Biotec) were equilibrated with BB, 0.4% BSA fol-

lowed by washing with BB. The column was placed inside the mag-

netic field of the SuperMACS II (Miltenyi Biotec), and the postnuclear

supernatant was loaded three times onto the column to maximize re-

covery. After washing with 20 ml of BB, the column was removed

from the magnetic field, and organelles associated with Fe beads

were serially eluted with 2 ml of BB containing either 100 mM or

300 mM KCl. Proteins were precipitated with 6.5% trichloroacetic

acid using 0.05% sodium deoxycholate as a carrier, solubilized,

and separated by SDS-NuPAGE (4%–12%; Invitrogen). Proteins

were silver stained, cut out, and after destaining, digested with tryp-

sin (Whitaker et al., 2004). Peptide mass fingerprinting was per-

formed using an ABI 4700 MALDI TOF-TOF (Applied Biosystems)

and the resulting sequences matched to the entries in the mouse

database. Some samples were analyzed by nano-LC MS/MS using

a quadrupole time-of-flight mass spectrometer (Micromass). For

Western blot analysis, the purification was carried out using MN cul-

tures derived from 16 rat embryos using buffers containing 100 mM

Na3VO4. Samples were separated by 10% SDS-PAGE.

Immunofluorescence and Axonal Retrograde Transport Assays

0.03–0.05 mg/ml of plasmids were microinjected into MN (DIV5–8) or

DRG (DIV2–4) as previously described (Deinhardt et al., 2006). Cells

were left to recover for 16–24 hr, unless otherwise indicated. MN

were then incubated with Alexa-TeNT HC (20–40 nM) or 0.2 mg/ml

Cy3-conjugated anti-p75NTR(EC), alone or together with 100 ng/ml

NGF or 40 nM TeNT HC, or partially purified BDNF-GFP (approxi-

mately 100 ng/ml) for 30 min at 37�C in Neurobasal, washed three

times with E4 supplemented with 30 mM HEPES-NaOH (pH 7.4),

and either fixed or imaged after 15 min at 37�C by time-lapse confo-

cal microscopy (Lalli and Schiavo, 2002). In some experiments, MN

were microinjected as described above, incubated with 40 nM Alexa

647-TeNT HC for 30 min and 30 nM TMRE for 5 min at 37�C before

washing with E4 containing 30 mM HEPES-NaOH (pH 7.4). For live

experiments using fluorescent TeNT HC-coupled Fe beads, MN

were first incubated with 80 nM Fe beads in HBSS, 0.2% BSA,

15 mg/ml polyornithine for 10 min at 4�C, washed with HBSS, and

left in complete medium for 20 min at 37�C. Cells were then washed

and imaged by time-lapse low-light microscopy. For internalization

assays, MN were incubated with biotinylated TeNT HC as described

previously (Deinhardt et al., 2006).

Image Analysis and Data Quantification

Images were acquired every 5 s over a total of up to 200 frames per

movie and analyzed as previously described (Lalli and Schiavo,

2002). Only moving carriers that could be tracked for at least four

time points were considered. The distance covered by a carrier be-

tween two consecutive frames (5 s), termed single movement, was

used to determine its instantaneous speed. A double-positive com-

partment was defined on the basis of the following criteria: (1) the

carrier was labeled in two different channels; (2) the morphology of

the carrier was very similar in the two channels; and (3) its speed

and direction were identical in the two channels for at least four

time points in a time series. Statistical analysis and curve fitting

were performed using Kaleidagraph (Synergy Software). Kymo-

graphs were generated using MetaMorph (Molecular Devices) after

rotation of the image stack to align the neuronal process vertically.

Horizontal single line-scans through the thickness of each process

were plotted sequentially for every frame in the time series.

Colocalization of TeNT HC and Rabs in fixed MN was quantified in

MetaMorph using the ‘‘manually count objects’’ option. For this, all

TeNT HC-positive structures were manually marked and automati-

cally counted, then the Rab channel was overlaid, and double-

positive structures were highlighted and counted. The p-CREB

mean intensity was measured in MetaMorph and is given in arbitrary

units. Measurements were performed in p-CREB-stained MN im-

aged in nonsaturating conditions and using the same area of interest

in the nuclei, stained with Draq5TM (Alexis). Student’s t test was

performed using Kaleidagraph (Synergy Software).

Supplemental Data

The Supplemental Data for this article can be found online at http://

www.neuron.org/cgi/content/full/52/2/293/DC1/.

Acknowledgments

We thank G.R.V. Hammond for the PH domain of cytohesin I; M.

Zerial (Max Planck-Institut, Dresden, D) for a Rab7 antibody; L.

Reichardt (University of California, San Francisco, CA) for the

chicken anti-TrkB antibody; V. Lessmann (Johannes Gutenberg

University, Mainz, D) for the rat pre-pro-BDNF-GFP construct;

M.V. Chao (Skirball Institute, New York University, New York, NY)

for the TrkB constructs; M. Koltzenburg and J. Crossley (Institute

of Child Health, London, UK) for help with the DRG cultures; and

M. Golding and A.P. Hibbert for advice. We are thankful to S. Tooze,

A. Behrens, and the Molecular NeuroPathobiology laboratory for

critical reading of the manuscript and to N. Totty for assistance

with the MS data. This work was supported by Cancer Research

UK, the European Molecular Biology Organization (S.S.), and Tele-

thon Italy (GGP05160 to C.B.).

Received: January 12, 2006

Revised: June 26, 2006

Accepted: August 7, 2006

Published: October 18, 2006

References

Adachi, H., Katsuno, M., Minamiyama, M., Sang, C., Pagoulatos, G.,

Angelidis, C., Kusakabe, M., Yoshiki, A., Kobayashi, Y., Doyu, M.,

et al. (2003). Heat shock protein 70 chaperone overexpression ame-

liorates phenotypes of the spinal and bulbar muscular atrophy trans-

genic mouse model by reducing nuclear-localized mutant androgen

receptor protein. J. Neurosci. 23, 2203–2211.

Bohnert, S., and Schiavo, G. (2005). Tetanus toxin is transported in

a novel neuronal compartment characterized by a specialized pH

regulation. J. Biol. Chem. 280, 42336–42344.

Bronfman, F.C., Tcherpakov, M., Jovin, T.M., and Fainzilber, M.

(2003). Ligand-induced internalization of the p75 neurotrophin re-

ceptor: a slow route to the signaling endosome. J. Neurosci. 23,

3209–3220.

Bucci, C., Parton, R.G., Mather, I.H., Stunnenberg, H., Simons, K.,

Hoflack, B., and Zerial, M. (1992). The small GTPase rab5 functions

as a regulatory factor in the early endocytic pathway. Cell 70, 715–

728.

Bucci, C., Thomsen, P., Nicoziani, P., McCarthy, J., and van Deurs,

B. (2000). Rab7: a key to lysosome biogenesis. Mol. Biol. Cell 11,

467–480.

Chavrier, P., Parton, R.G., Hauri, H.P., Simons, K., and Zerial, M.

(1990). Localization of low molecular weight GTP binding proteins

to exocytic and endocytic compartments. Cell 62, 317–329.

Claude, P., Hawrot, E., Dunis, D.A., and Campenot, R.B. (1982).

Binding, internalization, and retrograde transport of 125I-nerve

growth factor in cultured rat sympathetic neurons. J. Neurosci. 2,

431–442.

Deinhardt, K., Berninghausen, O., Willison, H.J., Hopkins, C.R., and

Schiavo, G. (2006). Tetanus toxin is internalized by a sequential cla-

thrin-dependent mechanism initiated within lipid microdomains and

independent of epsin1. J. Cell Biol. 174, 459–471.

Delcroix, J.D., Valletta, J.S., Wu, C.B., Hunt, S.J., Kowal, A.S., and

Mobley, W.C. (2003). NGF signaling in sensory neurons: evidence

that early endosomes carry NGF retrograde signals. Neuron 39,

69–84.

Page 12: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Neuron304

Devon, R.S., Orban, P.C., Gerrow, K., Barbieri, M.A., Schwab, C.,

Cao, L.P., Helm, J.R., Bissada, N., Cruz-Aguado, R., Davidson,

T.L., et al. (2006). Als2-deficient mice exhibit disturbances in endo-

some trafficking associated with motor behavioral abnormalities.

Proc. Natl. Acad. Sci. USA 103, 9595–9600.

Goldstein, L.S., and Yang, Z. (2000). Microtubule-based transport

systems in neurons: the roles of kinesins and dyneins. Annu. Rev.

Neurosci. 23, 39–71.

Grob, P.M., Ross, A.H., Koprowski, H., and Bothwell, M. (1985).

Characterization of the human melanoma nerve growth factor re-

ceptor. J. Biol. Chem. 260, 8044–8049.

Gruenberg, J., and Maxfield, F.R. (1995). Membrane transport in the

endocytic pathway. Curr. Opin. Cell Biol. 7, 552–563.

Gutierrez, M.G., Munafo, D.B., Beron, W., and Colombo, M.I. (2004).

Rab7 is required for the normal progression of the autophagic path-

way in mammalian cells. J. Cell Sci. 117, 2687–2697.

Guzik, B.W., and Goldstein, L.S. (2004). Microtubule-dependent

transport in neurons: steps towards an understanding of regulation,

function and dysfunction. Curr. Opin. Cell Biol. 16, 443–450.

Hadano, S., Benn, S.C., Kakuta, S., Otomo, A., Sudo, K., Kunita, R.,

Suzuki-Utsunomiya, K., Mizumura, H., Shefner, J.M., Cox, G.A., et al.

(2006). Mice deficient in the Rab5 guanine nucleotide exchange fac-

tor ALS2/alsin exhibit age-dependent neurological deficits and

altered endosome trafficking. Hum. Mol. Genet. 15, 233–250.

Hafezparast, M., Klocke, R., Ruhrberg, C., Marquardt, A., Ahmad-

Annuar, A., Bowen, S., Lalli, G., Witherden, A.S., Hummerich, H.,

Nicholson, S., et al. (2003). Mutations in dynein link motor neuron de-

generation to defects in retrograde transport. Science 300, 808–812.

Harrison, R.E., Bucci, C., Vieira, O.V., Schroer, T.A., and Grinstein, S.

(2003). Phagosomes fuse with late endosomes and/or lysosomes by

extension of membrane protrusions along microtubules: role of

Rab7 and RILP. Mol. Cell. Biol. 23, 6494–6506.

Holzbaur, E.L. (2004). Motor neurons rely on motor proteins. Trends

Cell Biol. 14, 233–240.

Houlden, H., King, R.H., Muddle, J.R., Warner, T.T., Reilly, M.M.,

Orrell, R.W., and Ginsberg, L. (2004). A novel RAB7 mutation associ-

ated with ulcero-mutilating neuropathy. Ann. Neurol. 56, 586–590.

Jager, S., Bucci, C., Tanida, I., Ueno, T., Kominami, E., Saftig, P., and

Eskelinen, E.L. (2004). Role for Rab7 in maturation of late autophagic

vacuoles. J. Cell Sci. 117, 4837–4848.

Johnson, E.M., Jr., Taniuchi, M., Clark, H.B., Springer, J.E., Koh, S.,

Tayrien, M.W., and Loy, R. (1987). Demonstration of the retrograde

transport of nerve growth factor receptor in the peripheral and cen-

tral nervous system. J. Neurosci. 7, 923–929.

Kanning, K.C., Hudson, M., Amieux, P.S., Wiley, J.C., Bothwell, M.,

and Schecterson, L.C. (2003). Proteolytic processing of the p75 neu-

rotrophin receptor and two homologs generates C-terminal frag-

ments with signaling capability. J. Neurosci. 23, 5425–5436.

Kieran, D., Kalmar, B., Dick, J.R., Riddoch-Contreras, J., Burnstock,

G., and Greensmith, L. (2004). Treatment with arimoclomol, a coin-

ducer of heat shock proteins, delays disease progression in ALS

mice. Nat. Med. 10, 402–405.

Lakadamyali, M., Rust, M.J., and Zhuang, X. (2006). Ligands for cla-

thrin-mediated endocytosis are differentially sorted into distinct

populations of early endosomes. Cell 124, 997–1009.

Lalli, G., and Schiavo, G. (2002). Analysis of retrograde transport in

motor neurons reveals common endocytic carriers for tetanus toxin

and neurotrophin receptor p75NTR. J. Cell Biol. 156, 233–239.

Lalli, G., Gschmeissner, S., and Schiavo, G. (2003). Myosin Va and

microtubule-based motors are required for fast axonal retrograde

transport of tetanus toxin in motor neurons. J. Cell Sci. 116, 4639–

4650.

McLauchlan, H., Newell, J., Morrice, N., Osborne, A., West, M., and

Smythe, E. (1998). A novel role for Rab5-GDI in ligand sequestration

into clathrin-coated pits. Curr. Biol. 8, 34–45.

Meckler, R.L., Baron, R., and McLachlan, E.M. (1990). Selective up-

take of C-fragment of tetanus toxin by sympathetic preganglionic

nerve terminals. Neuroscience 36, 823–829.

Perlson, E., Medzihradszky, K.F., Darula, Z., Munno, D.W., Syed,

N.I., Burlingame, A.L., and Fainzilber, M. (2004). Differential proteo-

mics reveals multiple components in retrogradely transported axo-

plasm after nerve injury. Mol. Cell. Proteomics 3, 510–520.

Pfeffer, S. (2003). Membrane domains in the secretory and endocytic

pathways. Cell 112, 507–517.

Puls, I., Jonnakuty, C., LaMonte, B.H., Holzbaur, E.L., Tokito, M.,

Mann, E., Floeter, M.K., Bidus, K., Drayna, D., Oh, S.J., et al.

(2003). Mutant dynactin in motor neuron disease. Nat. Genet. 33,

455–456.

Rink, J., Ghigo, E., Kalaidzidis, Y., and Zerial, M. (2005). Rab conver-

sion as a mechanism of progression from early to late endosomes.

Cell 122, 735–749.

Roux, S., Colasante, C., Saint Cloment, C., Barbier, J., Curie, T.,

Girard, E., Molgo, J., and Brulet, P. (2005). Internalization of a

GFP-tetanus toxin C-terminal fragment fusion protein at mature

mouse neuromuscular junctions. Mol. Cell. Neurosci. 30, 79–89.

Sakisaka, T., Meerlo, T., Matteson, J., Plutner, H., and Balch, W.E.

(2002). Rab-aGDI activity is regulated by a Hsp90 chaperone com-

plex. EMBO J. 21, 6125–6135.

Samuels, I.S., Seibenhener, M.L., Neidigh, K.B., and Wooten, M.W.

(2001). Nerve growth factor stimulates the interaction of ZIP/p62

with atypical protein kinase C and targets endosomal localization:

evidence for regulation of nerve growth factor-induced differentia-

tion. J. Cell. Biochem. 82, 452–466.

Saxena, S., Bucci, C., Weis, J., and Kruttgen, A. (2005a). The small

GTPase Rab7 controls the endosomal trafficking and neuritogenic

signaling of the nerve growth factor receptor TrkA. J. Neurosci. 25,

10930–10940.

Saxena, S., Howe, C.L., Cosgaya, J.M., Steiner, P., Hirling, H., Chan,

J.R., Weis, J., and Kruttgen, A. (2005b). Differential endocytic sorting

of p75NTR and TrkA in response to NGF: a role for late endosomes in

TrkA trafficking. Mol. Cell. Neurosci. 28, 571–587.

Schiavo, G., and van der Goot, F.G. (2001). The bacterial toxin tool-

kit. Nat. Rev. Mol. Cell Biol. 2, 530–537.

Snider, M.D. (2003). A role for rab7 GTPase in growth factor-regu-

lated cell nutrition and apoptosis. Mol. Cell 12, 796–797.

Stenmark, H., Parton, R.G., Steele-Mortimer, O., Lutcke, A., Gruen-

berg, J., and Zerial, M. (1994). Inhibition of rab5 GTPase activity

stimulates membrane fusion in endocytosis. EMBO J. 13, 1287–

1296.

Stockel, K., Schwab, M., and Thoenen, H. (1975). Comparison be-

tween the retrograde axonal transport of nerve growth factor and

tetanus toxin in motor, sensory and adrenergic neurons. Brain

Res. 99, 1–16.

Tucker, B.A., Rahimtula, M., and Mearow, K.M. (2005). A procedure

for selecting and culturing subpopulations of neurons from rat dor-

sal root ganglia using magnetic beads. Brain Res. Brain Res. Protoc.

16, 50–57.

Ure, D.R., and Campenot, R.B. (1997). Retrograde transport and

steady-state distribution of 125I-nerve growth factor in rat sympa-

thetic neurons in compartmented cultures. J. Neurosci. 17, 1282–

1290.

Verhoeven, K., De Jonghe, P., Coen, K., Verpoorten, N., Auer-Grum-

bach, M., Kwon, J.M., FitzPatrick, D., Schmedding, E., De Vriendt, E.,

Jacobs, A., et al. (2003). Mutations in the small GTPase late endoso-

mal protein RAB7 cause Charcot-Marie-Tooth type 2B neuropathy.

Am. J. Hum. Genet. 72, 722–727.

Vidricaire, G., and Tremblay, M.J. (2005). Rab5 and Rab7, but not

ARF6, govern the early events of HIV-1 infection in polarized human

placental cells. J. Immunol. 175, 6517–6530.

Vonderheit, A., and Helenius, A. (2005). Rab7 associates with early

endosomes to mediate sorting and transport of Semliki forest virus

to late endosomes. PLoS Biol. 3, e233. 10.1371/journal.pbio.

0030233.

Whitaker, H.C., Hanrahan, S., Totty, N., Gamble, S.C., Waxman, J.,

Cato, A.C., Hurst, H.C., and Bevan, C.L. (2004). Androgen receptor

is targeted to distinct subcellular compartments in response to dif-

ferent therapeutic antiandrogens. Clin. Cancer Res. 10, 7392–7401.

Wilson, J.M., de Hoop, M., Zorzi, N., Toh, B.H., Dotti, C.G., and

Parton, R.G. (2000). EEA1, a tethering protein of the early sorting

Page 13: Rab5 and Rab7 Control Endocytic Sorting along the Axonal Retrograde Transport Pathway

Rab5 and Rab7 Regulate Axonal Transport305

endosome, shows a polarized distribution in hippocampal neurons,

epithelial cells, and fibroblasts. Mol. Biol. Cell 11, 2657–2671.

Yan, Q., Elliott, J.L., Matheson, C., Sun, J., Zhang, L., Mu, X., Rex,

K.L., and Snider, W.D. (1993). Influences of neurotrophins on mam-

malian motoneurons in vivo. J. Neurobiol. 24, 1555–1577.

Zerial, M., and McBride, H. (2001). Rab proteins as membrane orga-

nizers. Nat. Rev. Mol. Cell Biol. 2, 107–117.

Zhang, Y., Moheban, D.B., Conway, B.R., Bhattacharyya, A., and

Segal, R.A. (2000). Cell surface Trk receptors mediate NGF-induced

survival while internalized receptors regulate NGF-induced differen-

tiation. J. Neurosci. 20, 5671–5678.