ArticlePDF Available

Transactivation of Trk Neurotrophin Receptors by G-Protein-Coupled Receptor Ligands Occurs on Intracellular Membranes

Authors:

Abstract and Figures

Neurotrophins, such as NGF and BDNF, activate Trk receptor tyrosine kinases through receptor dimerization at the cell surface followed by autophosphorylation and intracellular signaling. It has been shown that activation of Trk receptor tyrosine kinases can also occur via a G-protein-coupled receptor (GPCR) mechanism, without involvement of neurotrophins. Two GPCR ligands, adenosine and pituitary adenylate cyclase-activating polypeptide (PACAP), can activate Trk receptor activity to increase the survival of neural cells through stimulation of Akt activity. To investigate the mechanism of Trk receptor transactivation, we have examined the localization of Trk receptors in PC12 cells and primary neurons after treatment with adenosine agonists and PACAP. In contrast to neurotrophin treatment, Trk receptors were sensitive to transcriptional and translational inhibitors, and they were found predominantly in intracellular locations particularly associated with Golgi membranes. Biotinylation and immunostaining experiments confirm that most of the transactivated Trk receptors are found in intracellular membranes. These results indicate that there are alternative modes of activating Trk receptor tyrosine kinases in the absence of neurotrophin binding at the cell surface and that receptor signaling may occur and persist inside of neuronal cells.
Content may be subject to copyright.
Cellular/Molecular
Transactivation of Trk Neurotrophin Receptors by
G-Protein-Coupled Receptor Ligands Occurs on
Intracellular Membranes
Rithwick Rajagopal,
1
Zhe-Yu Chen,
2
Francis S. Lee,
2
and Moses V. Chao
1
1
Molecular Neurobiology Program, Skirball Institute of Biomolecular Medicine, Departments of Cell Biology and Physiology and Neuroscience, New York
University School of Medicine, New York, New York 10016, and
2
Department of Psychiatry, Weill Medical College of Cornell University, New York, New
York 10021
Neurotrophins, such as NGF and BDNF, activate Trk receptor tyrosine kinases through receptor dimerization at the cell surface followed
by autophosphorylation and intracellular signaling. It has been shown that activation of Trk receptor tyrosine kinases can also occur via
a G-protein-coupled receptor (GPCR) mechanism, without involvement of neurotrophins. Two GPCR ligands, adenosine and pituitary
adenylate cyclase-activating polypeptide (PACAP), can activate Trk receptor activity to increase the survival of neural cells through
stimulation of Akt activity. To investigate the mechanism of Trk receptor transactivation, we have examined the localization of Trk
receptors in PC12 cells and primary neurons after treatment with adenosine agonists and PACAP. In contrast to neurotrophin treatment,
Trk receptors were sensitive to transcriptional and translational inhibitors, and they were found predominantly in intracellular locations
particularly associated with Golgi membranes. Biotinylation and immunostaining experiments confirm that most of the transactivated
Trk receptors are found in intracellular membranes. These results indicate that there are alternative modes of activating Trk receptor
tyrosine kinases in the absence of neurotrophin binding at the cell surface and that receptor signaling may occur and persist inside of
neuronal cells.
Key words: NGF; tyrosine phosphorylation; basal forebrain; adenosine; PACAP; Golgi apparatus
Introduction
Neurotrophins are essential regulators of neuronal development,
growth, and differentiation in the vertebrate nervous system
(Huang and Reichardt, 2001). The actions of neurotrophins are
dictated by two classes of cell surface receptors, the Trk receptor
tyrosine kinase and the p75 neurotrophin receptor, a member of
the tumor necrosis factor (TNF) receptor superfamily (Hemp-
stead, 2002; Chao, 2003a; Huang and Reichardt, 2003). Although
NGF binds exclusively to TrkA, BDNF and neurotrophin-4
(NT-4) bind to TrkB, and NT-3 binds to TrkC, all neurotrophins
bind to p75. After binding, neurotrophins and each of their re-
ceptors undergo internalization and transport from axon termi-
nals to neuronal cell bodies (Hendry et al., 1974; DiStefano et al.,
1992; Bronfman et al., 2003), in which a number of transcrip-
tional and enzymatic activities are activated. Engagement of Trk
receptors results in increases in cAMP response element-binding
protein (CREB) and extracellular signal-related kinase 5 (Erk5)
activities (Riccio et al., 1997; Watson et al., 1999b, 2001), as well
as phosphoinositide lipid phosphorylation and activation of GT-
Pases, such as Ras and Rap1 (York et al., 1998).
Another mode of receptor activation is through transactiva-
tion of receptor tyrosine kinases in response to G-protein-
coupled receptor signaling (Daub et al., 1996; Luttrell et al., 1999;
Fischer et al., 2003). For example, receptors for epidermal growth
factor (EGF), insulin-like growth factor-1, and platelet-derived
growth factor can be transactivated through G-protein-coupled
receptors to give proliferative and mitogen-activated protein ki-
nase (MAPK) responses. Also, signaling through dopamine re-
ceptors leads to PDGF receptor activation that results in changes
in synaptic transmission (Kotecha et al., 2002). Hence, transacti-
vation can lead to effects on cell proliferation, differentiation, and
synaptic plasticity; however, the cellular mechanisms that ac-
count for these transactivation effects are not well understood.
We reported previously that TrkA and TrkB receptors can be
activated by ligands of the G-protein-coupled receptor (GPCR)
family of transmembrane receptors in the absence of NGF or
BDNF (Lee and Chao, 2001; Lee et al., 2002b). The ligands in-
clude the nucleoside adenosine or adenosine agonists, such as
CGS 21680, and the neuropeptide pituitary adenylate cyclase-
activating polypeptide (PACAP). Adenosine and CGS 21680 rec-
ognize the A
2A
receptor (Jarvis et al., 1989), whereas PACAP
binds to the PAC1 receptor (Ishihara et al., 1992). Not only do
adenosine and PACAP activate Trk receptors, but effectors of the
Received Jan. 2, 2004; revised June 8, 2004; accepted June 14, 2004.
This work was supported by National Institutes of Health Grants NS21072 and HD23315 (M.V.C.) and the White-
hall Foundation and the National Alliance for Research on Schizophrenia and Depression (F.S.L.). We thank Juan
Carlos Arevalo for generating Trk antibodies, Lani Mustacchi for primary cultures, Daniela Pereira for advice, and
Wen-biao Gan for advice on confocal microscopy.
Correspondence should be addressed to Moses V. Chao, Skirball Institute of Biomolecular Medicine, New York
University School of Medicine, 540 First Avenue, New York, NY 10016. E-mail: chao@saturn.med.nyu.edu.
DOI:10.1523/JNEUROSCI.0010-04.2004
Copyright © 2004 Society for Neuroscience 0270-6474/04/246650-09$15.00/0
6650 The Journal of Neuroscience, July 28, 2004 24(30):6650 6658
Trk tyrosine kinase are also phosphorylated in response to these
ligands. Adenosine treatment of PC12 cells promotes the phos-
phorylation of Shc adaptor proteins, as well as phospholipase C
,
analogous to the induction observed with NGF. Activation of
phosphoinositide (PI)-3 kinase and Akt accounted for neuropro-
tective effects afforded by transactivation by adenosine and
PACAP. The effects of adenosine and PACAP can be specifically
blocked by K252a, an inhibitor of Trk tyrosine kinases.
In the present study, we report that Trk activation by adeno-
sine and its agonist CGS 21680 or PACAP occurs exclusively in an
intracellular location and partly involves the Golgi apparatus.
Biotinylation assays indicate that CGS 21680 activates an intra-
cellular pool of Trk receptors. Indirect immunofluorescence ex-
periments confirm that only intracellular Trk receptors are acti-
vated by CGS 21680. Furthermore, colocalization of activated
Trk receptors and markers of the Golgi apparatus, such as
TGN-38 and GM130, suggest that Trk receptors can reside and be
activated in Golgi membranes. Taken together with previous ob-
servations, these results suggest that intracellular membranes
may potentially serve as a platform for Trk receptors to mediate
cell survival through the PI3K–Akt pathway.
Materials and Methods
Reagents. Adenosine and CGS 21680 were obtained from Sigma (St.
Louis, MO). PACAP 38 was from Peninsula Laboratories (San Carlos,
CA). K252a and brefeldin A were purchased from Calbiochem (La Jolla,
CA). Cycloheximide and actinomycin D were purchased from Sigma.
Sulfo-NHS-LC-biotin, sulfo-NHS-acetate, and streptavidin-agarose
were purchased from Pierce Biotechnology (Rockford, IL). NGF was
obtained from Harlan Bioproducts (Indianapolis, IN), and BDNF was
from PeproTech (Rocky Hill, NJ). Anti-pan-Trk rabbit antiserum raised
against the C-terminal region of the Trk receptor was from Barbara
Hempstead (#45, Weill Medical College, New York, NY), and C-14 pan-
Trk was from Santa Cruz Biotechnology (Santa Cruz, CA). Mouse
monoclonal anti-Trk antibodies against the C terminus of Trk were ob-
tained from Santa Cruz Biotechnology (B-3), and the TrkE7 antibody
against the extracellular domain of TrkA was from Zymed (San Fran-
cisco, CA). Phosphotyrosine antibodies (pY99), monoclonal Akt anti-
bodies (Akt B-1), and Erk 1/2 antibodies were from Santa Cruz Biotech-
nology. Antibodies to Y490 and Y674/Y675 of Trk as well as antibodies to
phosphorylated Akt (pAkt S473) and phosphorylated ERK1/2 were pur-
chased from Cell Signaling Technology (Beverly, MA). Mouse monoclo-
nal antibodies to Golgi matrix 130 (GM130) and early endosome
antigen-1 (EEA-1) were from BD Biosciences (San Diego, CA). Mouse
monoclonal antibodies to trans Golgi network-38 (TGN-38) were from
Affinity Bioreagents (Golden, CO).
Immunoprecipitation and immunoblotting. PC12-615 cells (Hemp-
stead et al., 1992) were maintained in DMEM containing 5% fetal bovine
serum, 10% horse serum supplemented with 2 mMglutamine plus 250
g/ml G418. For all experiments, cells were serum-starved by washing
twice with PBS and placing in serum-free media for 24 hr. In some cases,
cells were pretreated with 5
g/ml brefeldin A, 20
g/ml cycloheximide,
200 ng/
l actinomycin D, or vehicle. Cell lysates from PC12 cells were
prepared by incubating in lysis buffer (1% Nonidet P-40, 150 mMNaCl,
1m
MEDTA, 10 mMTris, pH 8.0, 10% glycerol, 10 mMNaF, 1 mMsodium
orthovanadate, 2
g/ml aprotinin, 1
g/ml leupeptin, and 25
g/ml
phenylmethylsulfonyl fluoride) for 30 min on ice. Clarified lysates were
immunoprecipitated by incubating overnight at 4°C with anti-pan-Trk
polyclonal antibody (Trk C-14) followed by incubation with protein
A-Sepharose beads. Equivalent amounts of protein were analyzed for
each condition. The beads were washed three times with lysis buffer, and
the immune complexes were boiled in SDS-sample buffer and loaded on
SDS-PAGE gels for immunoblot analysis. The immunoreactive protein
bands were detected by enhanced chemiluminescence (Amersham Bio-
sciences, Arlington Heights, IL).
RT-PCR analysis. Total RNA from PC12-615 cells or from subman-
dibular gland dissected from adult Sprague Dawley rats was isolated
using TRIZOL (Invitrogen, Carlsbad, CA). Five hundred nanograms of
total RNA were subjected to reverse transcription using avian myeloblas-
tosis virus reverse transcriptase (Roche Applied Science, Penzberg, Ger-
many) for 30 min at 50°C. Total RNA not incubated with reverse tran-
scriptase was used as a negative control. cDNA products were incubated
with TaqDNA polymerase (Roche Applied Science) and primers for rat
NGF or for glyceraldehyde-3-phosphate dehydrogenase (GAPDH).
Primers used for NGF amplification were designed as described previ-
ously (Donovan et al., 1995): NGF, 5-CGCTCATCCACCCACCCAGT-
CTTC, 3-CTTGACAAAGGTGTGAGTCGTGGT. These primers
yielded a product of 267 base pairs. After an initial 95°C hot start, 35
cycles of 94°C for 30 sec, 60°C for 1 min, and 68°C for 1 min were
performed, followed by a final extension of 68°C for 5 min. Products
were resolved by electrophoresis using a 2% agarose gel and visualized
using ethidium bromide.
Cell surface biotinylation. Serum-starved cells were rinsed twice with
PBS containing 1 mMCaCl
2
and 0.5 mMMgCl
2
(PBS-Ca-Mg) and then
incubated with 0.5 mg/ml sulfo-NHS-LC-biotin dissolved in biotinyla-
tion buffer (0.01 MTEA, pH 7.4, 2 mMCaCl
2
, 150 mMNaCl) for 30 min
at 4°C. Cells were then quenched with PBS-Ca-Mg containing 100 mM
glycine for 20 min at 4°C. After warming to 37°C, cells were treated with
10 nMCGS 21680, 100 ng/ml NGF, or vehicle media for the indicated
times. Cells were then lysed in RIPA buffer (1% NP-40, 0.1% SDS, 0.1%
deoxycholate, 150 mMNaCl, 1 mMEDTA, 10 mMTris, pH 8.0, 10 mM
NaF, 1 mMsodium orthovanadate, 2
g/ml aprotinin, 1
g/ml leupeptin,
and 25
g/ml phenylmethylsulfonyl fluoride) for 30 min on ice. Clarified
lysates were then split into two equal portions, one that was immunopre-
cipitated with streptavidin-agarose and the other with Trk C-14.
Receptor insertion assay. Serum-starved PC12 cells were rinsed twice
with PBS-Ca-Mg and incubated with 1.5 mg/ml sulfo-NHS-acetate in
blocking buffer (0.1 Msodium phosphate buffer, pH 7.4, 150 mMNaCl)
for 30 min at 4°C. The blocking reaction was quenched for 20 min at 4°C
using 100 mMglycine dissolved in PBS-Ca-Mg. Cells were subsequently
allowed to warm to 37°C and then treated with CGS 21680 or NGF as
indicated. After treatment, surface biotinylation and lysis were per-
formed as outlined above. Cell lysates were subjected to immunoprecipi-
tation with streptavidin-agarose. Material obtained from this immuno-
precipitation was labeled “cell surface pool.” Supernatants from the
initial immunoprecipitation were subjected to a second immunoprecipi-
tation with streptavidin-agarose to clear the lysate of biotinylated mate-
rial. The second supernatant was confirmed to be free of biotinylated
proteins by blotting with streptavidin-HRP. TrkA was then immunopre-
cipitated from the biotin-cleared lysate using Trk C-14 antibodies and
labeled “internal pool.” Immunoprecipitates were analyzed by blotting
with phosphotyrosine antibodies, phospho-Trk antibodies, or pan-Trk
antibodies.
Generation and affinity purification of antibodies against phosphorylated
TrkA. A peptide containing phosphorylated tyrosine 794 of rat TrkA,
previously shown to be a site for phospholipase C-
(PLC-
) recruitment
in response to NGF (Loeb et al., 1994), was used to immunize rabbits
(LQALAQAPPSpYLDVC). Crude serum was first passed through an af-
finity column made by coupling the nonphosphorylated version of the
peptide to cyanogen bromide-activated Sepharose beads. Flow through
from this column was then passed though a column made by coupling
the phospho-peptide to cyanogen bromide-activated Sepharose. The col-
umn was washed extensively with Tris-buffered saline containing 0.1%
Tween 20, and antibodies were then eluted with 0.1 Mglycine, pH 2.5.
Fractions were collected from the eluate, neutralized with Tris buffer, pH
9.5, and then tested for specificity by blotting NGF-treated PC12 cell
lysates. Specificity was further confirmed by a peptide competition assay
in which a fivefold weight excess of either phosphorylated or nonphos-
phorylated TrkA peptide was preincubated with antibody for 2 hr at 4°C
and then used for Western blotting.
Basal forebrain cell cultures. Dissociated primary cultures of basal fore-
brain neurons from embryonic day 18 (E18) rats were prepared from
timed-pregnant Sprague Dawley rats as described previously (Aibel et al.,
1998). Fetuses were removed under sterile conditions and kept in PBS on
ice for microscopic dissection of the basal forebrain. The meninges were
removed, and the tissue was placed in HBSS supplemented with 0.37%
Rajagopal et al. Activation of Trk Receptors J. Neurosci., July 28, 2004 24(30):6650 6658 6651
glucose. The tissue was minced briefly with fine forceps and then tritu-
rated with a fire-polished Pasteur pipette. Cells were counted and plated
on culture wells coated with 0.01 mg/ml poly-L-lysine overnight in Neu-
robasal media containing B27 supplement and L-glutamine (0.5 mM).
Experiments were conducted 10 d after plating.
Immunofluorescence and confocal microscopy. Cells were cultured on
Nunc Permanox eight-well chamber slides coated with poly-L-lysine.
Twenty-four hours after plating, cells were serum-starved by washing
twice with PBS and placing in serum-free media for 24 hr. Cells were
fixed directly after treatment in 4% paraformaldehyde for 10 min at
room temperature. In some cases, cells were pretreated with 5
g/ml
brefeldin A. After two rinses in PBS, cells were permeabilized with 0.1%
Triton X-100 for 10 min on ice. Cells were blocked in 5% BSA/PBS for 30
min at room temperature before incubation for 3 hr at room temperature
with primary antibody diluted in blocking buffer. Cells were washed
three times with PBS and then incubated with secondary antibody
(FITC-conjugated anti-rabbit; Cy3-conjugated anti-mouse; Jackson Im-
munoResearch, West Grove, PA; 1:100) diluted in blocking buffer for 1
hr at room temperature. Cells were washed three times in PBS and then
mounted in Vectashield mounting medium containing 4,6-diamidino-
2-phenylindole (DAPI). Standard fluorescence microscopy was per-
formed using a Nikon Eclipse E800 microscope equipped with a Nikon
Plan Apo 60, 1.4 numerical aperture (NA) oil immersion objective.
Images were acquired with a Zeiss AxioCam HRc digital camera. Confo-
cal images were acquired using a Bio-Rad Radiance 2000 confocal unit
coupled to an Olympus BX5OWI microscope equipped with an Olym-
pus PlanApo 60, 1.4 NA oil immersion objective.
Results
GPCR ligands activate TrkA isoforms
Treatment of hippocampal neurons or
PC12 cells with adenosine resulted in an
activation of Trk activity and increased cell
survival in the absence of BDNF or NGF
(Lee and Chao, 2001). Transactivation of
Trk neurotrophin receptors leads to selec-
tive induction of the PI3-kinaseAkt path-
way that accounts for the survival re-
sponse. The increase in Trk activity was
inhibited by protein kinase inhibitors,
such as PP1, specific for the Src family
members. The Trk-dependent activation
of Akt could be blocked with LY294002, a
PI3-kinase inhibitor. PACAP also transac-
tivated Trk receptors in a manner very
similar to adenosine (Lee et al., 2002b), al-
though many other GPCR ligands failed to
transactivate Trk receptors.
To determine which residues of the Trk
receptor are phosphorylated after GPCR
stimulation, we performed phosphoryla-
tion analysis of Trk in PC12-615 cells
(Hempstead et al., 1992). These cells ex-
press elevated levels of TrkA and have been
widely used to study signal transduction.
TrkA was immunoprecipitated from cells
treated with 10
Madenosine and ana-
lyzed with antibodies to specific Trk phos-
phorylation sites, including the activation
loop of the kinase domain phosphoty-
rosines (amino acids 683 and 684 of rat
TrkA) and the Shc-binding phosphoty-
rosine site (amino acid 490). Indeed, all of
these tyrosine residues were phosphory-
lated in response to adenosine treatment
(Fig. 1A). The increase in specific phos-
phorylation sites is consistent with the finding that Trk enzymatic
activity is enhanced after adenosine or PACAP treatment (Lee et
al., 2002b). Furthermore, the phosphorylation events occurred in
a similar time interval for all three residues, which involved at
least 60 min to observe the activation of TrkA.
Phosphorylation of the activation-loop and Shc-binding res-
idues was observed in both the 140 and 110 kDa isoforms of Trk.
The phosphotyrosine analysis clearly indicated that the imma-
ture 110 kDa form of Trk was activated first, followed by the
mature 140 kDa form (Fig. 1A). The Trk receptor exists in a
number of different glycosylation states within the cell, but only
the fully glycosylated 140 kDa receptor is thought to be targeted
to the plasma membrane (Watson et al., 1999a; Jullien et al.,
2002). At the cell surface, NGF engages only the fully mature
receptor found to initiate signaling. The appearance of immature,
underglycosylated forms of TrkA early in the time course of
transactivation suggests that intracellular processing events may
play an important role in the mechanism and that a distinct pool
of Trk receptors is involved in this process.
To assess whether the activation and processing of Trk recep-
tors depended on transcriptional or posttranscriptional events,
we performed analyses using inhibitors of transcription and
translation. PC12 cells were pretreated with either actinomycin D
(0.2
g/
l) or cycloheximide (20
g/ml) for 30 min and then
Figure 1. Adenosine activates TrkA receptors at multiple phosphotyrosine residues. A, PC12 cells were treated with 10
Madenosine for the indicated times. Trk was immunoprecipitated from cell lysates using a pan-Trk antibody. Phosphor-
ylation of Trk was assessed with antibodies against phosphotyrosine 499 of rat TrkA [pTrk 490 (Cell Signaling)] or
antibodies against phosphotyrosines 683 and 684 of rat TrkA [pTrk 674/675 (Cell Signaling)]. Immunoblot with pan-Trk
antibody (B3) was used to ensure equal loading. B, PC12 cells were pretreated with either 20
g/ml cycloheximide or 200
ng/
l actinomycin D for 30 min before incubation with 10 nMCGS 21680 for 1 or 3 hr. Trk was immunoprecipitated from
cell lysates and analyzed with phosphotyrosine antibodies. Trk protein levels were determined by blotting with pan-Trk
antisera. C, Quantitation of phospho-Trk and total Trk levels from lysates of CGS 21680-treated PC12-615 cells. Three
independent experiments were quantified using ImageJ analysis software. Levels of band intensity relative to untreated
control are represented on the y-axis. Error bars reflect SEM. D, RT-PCR for NGF mRNA was performed on total RNA
extracted from untreated PC12 cells or cells treated with 10 nMCGS 21680 for 3 hr. RNA extracted from adult rat subman-
dibular gland was used as a positive control; GAPDH was used as an internal loading control.
6652 J. Neurosci., July 28, 2004 24(30):6650 6658 Rajagopal et al. Activation of Trk Receptors
treated with CGS 21680 (10 nM). CGS
21680 was used instead of adenosine be-
cause it is a specific and stable agonist that
interacts with the A
2A
receptor (Jarvis et
al., 1989). Previous experiments indicated
that CGS 21680 elicited identical re-
sponses as adenosine (Lee et al., 2001).
Phosphorylation of Trk was assessed by
immunoprecipitation from lysates using a
pan-Trk antibody followed by blotting
with a phosphotyrosine antibody, pY99.
After 1 and 3 hr of CGS 21620 treatment,
we consistently observed phosphorylation
of the 110 kDa TrkA species. Pretreatment
with actinomycin D, a potent inhibitor of
cellular transcription, or cycloheximide, a
translation inhibitor, prevented the trans-
activation of Trk by the adenosine agonist
CGS 21680 (Fig. 1B). Hence, GPCR-
mediated transactivation of Trk receptors
requires transcriptional and protein syn-
thesis events that may influence the pro-
cessing and activation of the receptor. An
increase in Trk receptor levels was ob-
served after 3 hr of treatment with CGS
21680; however, it should be noted that acti-
vation of TrkA occurred before this increase.
Quantitation of multiple experiments clearly
indicates that TrkA activity was induced af-
ter 1 hr of CGS 21680 treatment (Fig. 1C), a
time at which TrkA receptors were not sig-
nificantly upregulated.
To determine whether the gene up-
regulation requirement for transactivation
directly involved production of NGF, we
performed RT-PCR analysis on PC12 cells
treated with CGS 21680. As shown in Fig-
ure 1D, NGF mRNA was not detected in
either untreated or CGS-treated PC12
cells. In contrast, NGF mRNA was readily
detected in adult rat submandibular gland.
Previous experiments also failed to detect
any NGF biological activity in cells treated
with GPCR ligands (Lee et al., 2002).
Therefore, it is unlikely that transactiva-
tion of TrkA receptors involves NGF.
GPCR ligands activate an internal pool of Trk receptors
The finding that immature forms of Trk were activated preferen-
tially early during the time course of transactivation (Fig. 1A)
suggested than an intracellular pool Trk receptors is targeted. To
localize the site of Trk receptor transactivation, we performed a
cell surface biotinylation procedure, as outlined in Figure 2A.
PC12 cells were subjected to surface labeling with sulfo-NHS-
biotin and then treated with 10 nMCGS 21680 or 100 ng/ml NGF.
After 10 min of NGF treatment, a considerable amount of
activated TrkA receptors could be found in the biotinylated cell
surface pool of proteins (Fig. 2 B); however, at no point after CGS
21680 treatment was phosphorylated TrkA detected in the cell
surface pool, although transactivated Trk appeared in the total
Trk pool after 2 hr of CGS 21680 treatment (Fig. 2B). This de-
layed time course was consistent with previous observations (Lee
and Chao, 2001). The lack of receptors in the biotinylated frac-
tion suggests that most of the transactivated receptors did not
arise from the cell surface pool.
Although these biotinylation results revealed that preexisting
surface Trk receptors were not involved in the transactivation
mechanism, the possibility still remained that newly synthesized
Trk receptors induced by GPCR activity could be inserted into
the plasma membrane and become activated. To assess whether
the newly synthesized Trk receptors are correctly processed and
inserted into the cell membrane after CGS 21680 treatment, we
performed a membrane receptor insertion assay using a modified
cell surface biotinylation procedure (Peng et al., 2001). Cells were
first blocked from biotinylation using sulfo-NHS-acetate and
then treated with 10 nMCGS 21680 for the indicated times (Fig.
3A). After treatment, cells were biotinylated with sulfo-NHS-LC-
biotin and then subjected to immunoprecipitation. Nonbiotiny-
lated Trk was isolated by immunoprecipitation with pan-Trk an-
tibodies from the biotin-depleted lysate.
Figure 2. Transactivated Trk receptors are not detected at the cell surface. A, A schematic diagram of the biotinylation
procedureisshown.PC12 cells were biotinylated as described in Materials andMethodsandthentreated with 10 nMCGS 21680 for
the indicated times or with 100 ng/ml NGF for 10 min. Cell lysates were prepared and split into two equal portions. Half of the
lysate was subjected to immunoprecipitation with streptavidin-agarose, and the other half was immunoprecipitated with pan-
Trk antibodies. B, Immunoprecipitates were analyzed by blotting with phosphotyrosine antibodies and with pan-Trk antibodies.
Figure 3. TrkAistransactivatedon internal membranes. A, A receptor insertion assay wasperformedasdescribed in Materials
and Methods and is depicted in the schematic diagram. PC12 cells were treated with the biotinylation-blocking reagent S-NHS-
acetate and then treated with either 10 nMCGS 21680 or 100 ng/ml NGF for 3 hr. Cell lysates were immunoprecipitated with
avidin-agarose. Supernatants from the avidin-agarose immunoprecipitation that were depleted of biotinylated material were
subsequently subjected to immunoprecipitation with pan-Trk antibodies. B, Immunoprecipitates were analyzed by blotting with
phosphotyrosine antibodies, antibodies to phosphorylated Trk, or pan-Trk antibodies. Untreated cells that were biotinylated
immediately after block (lane 1) or 3 hr after block (lane 2) were used as controls for basal levels.
Rajagopal et al. Activation of Trk Receptors J. Neurosci., July 28, 2004 24(30):6650 6658 6653
This procedure allowed us to measure the newly inserted
membrane proteins during a treatment period after the blocking
step. The level of receptor insertion was then determined by sub-
sequent SDS-PAGE and immunoblotting with pan-Trk antibod-
ies. The basal level of receptor insertion was determined by com-
paring cells that were blocked and then immediately biotinylated
(Fig. 3B, lane 1) with cells that were blocked and left in serum-free
media for 3 hr before biotinylation (lane 2). All conditions of
treatment were compared with cells that were left untreated for 3
hr (lane 2).
Treatment with NGF gave rise to a detectable increase in acti-
vated Trk receptors at the cell surface (Fig. 3B, lane 7). An in-
crease in the amount of biotinylated Trk was observed with in-
creasing periods of incubation with CGS 21680 (top panel).
Insertion of receptors can be seen as early as after 1 hr of treat-
ment (lane 4), and maximal insertion is detected at 3 hr of treat-
ment (lane 6). Notably, NGF did not induce significant Trk re-
ceptor insertion after 3 hr of treatment (Fig. 3B, compare lane 7
and lane 2).
To determine whether newly inserted Trk receptors induced
by CGS 21680 treatment are activated, we reblotted the top panel
of Figure 3Bwith a phosphotyrosine antibody (pY). Surprisingly,
none of the Trk receptors at the cell surface were tyrosine phos-
phorylated after treatment with CGS 21680. After NGF treat-
ment, however, surface receptors were robustly activated. These
results were confirmed by blotting with an antibody specific to
phosphotyrosine 490 site of Trk (pTrk).
These results indicate that the activation of Trk receptors by
CGS 21680 is restricted to an internal pool of receptors that do
not reach the cell membrane. To confirm that Trk receptors
transactivated by CGS 21680 are found inside the cell, we ana-
lyzed the nonbiotinylated lysate by phosphotyrosine blotting. In-
deed, activation of TrkA by CGS 21680 was detected in the inter-
nal pool of receptors after 2 hr (Fig. 3B, bottom panel), which is
consistent with previous measurements with adenosine and
PACAP (Lee et al., 2001, 2002b).
Immunostaining with phospho-specific TrkA antibody
Previous experiments in PC12 cells and sympathetic neurons in
compartmentalized cultures have provided evidence that Trk re-
ceptor signaling occurs in intracellular locations (Grimes et al.,
1997; Tsui-Pierchala and Ginty, 1999; Jullien et al., 2002; Ye et al.,
2003). To address the intracellular localization of transactivated
Trk receptors, we subjected PC12 cells to indirect immunofluo-
rescence to determine the intracellular location of Trk activation
by GPCR ligands.
To follow activated Trk receptors, we generated a new
phospho-specific TrkA antibody for this purpose. The antibody
was made against a phosphopeptide spanning the PLC-
site at
Y785. Western blot analysis of the affinity-purified antibody
showed that it specifically recognized the activated TrkA receptor
and not the unstimulated receptor in PC12 cells (Fig. 4). Speci-
ficity for the phosphorylated form of TrkA was confirmed using
peptide competition assays. As shown in Figure 4, the phos-
phopeptide, and not the unphosphorylated peptide, effectively
blocked binding of the pTrkA antibody to immunoblots from
NGF-treated PC12 cell lysates. This antibody did not recognize
TrkB receptors in transfected and hippocampal cells or other
tyrosine phosphorylated proteins (data not shown).
To image TrkA receptors by immunofluorescent microscopy,
we serum-starved PC12 cells for 24 hr before treatment with
either CGS 21680 or 100 ng/ml NGF for 5 min. Cells were fixed,
permeabilized, and stained with the phospho-TrkA-specific an-
tibody generated against the C-terminal phosphotyrosine. Cells
were simultaneously costained with pan-Trk antibodies. As seen
in Figure 5, untreated PC12 cells display Trk localization that
spans the entire cell. Trk (red) was detected uniformly at the
plasma membrane as well as in numerous small-diameter intra-
cellular vesicles and in a perinuclear compartment; however, un-
treated cells display only a negligible amount of phospho-Trk
immunoreactivity (pTrkA) that is mostly limited to the small
intracellular vesicles. On the other hand, cells treated with NGF
for 5 min displayed punctate staining for phospho-Trk along the
plasma membrane but not within the cell, where there is an ex-
tensive amount of nonactivated Trk as shown by the pan-Trk
staining in red.
The staining pattern of phospho-Trk that is observed after
CGS 21680 treatment is quite distinct from that observed in
NGF-treated cells. As seen in Figure 5A(bottom panels),
phospho-Trk staining in cells treated with CGS 21680 for 3 hr was
concentrated to closely apposed large vesicles in the perinuclear
region of the cell. Staining was absent along the plasma mem-
brane, consistent with our findings that GPCR-activated Trk is
limited to an intracellular pool of receptors.
The staining pattern that we observed was reminiscent of
Golgi staining from PC12 cells (Kim and von Zastrow, 2003). To
confirm the Golgi localization, we analyzed PC12 cells treated
with CGS 21680 by confocal microscopy using anti-phospho-Trk
(green) and antibodies against GM130 or TGN38 (red), both of
which are well established markers of the Golgi apparatus (Luzio
et al., 1990; Nakamura et al., 1995). As shown, phospho-Trk par-
tially colocalizes with both of these Golgi markers (Fig. 5B). On
the other hand, cells stained with a marker for early endosomes,
EEA-1 (Fig. 5B), or for late endosomes, LAMP1, displayed much
less colocalization with phospho-Trk after GPCR stimulation
(data not shown). These results suggest that GPCR-activated Trk
is restricted to an intracellular region that is not associated with
the endocytic trafficking pathway and suggests that part of the
activated pool is detected close to or contiguous with the Golgi
apparatus.
Brefeldin A treatment
To verify that the Golgi is involved in transactivation by GPCRs,
we disrupted cells with brefeldin A, a fungal metabolite that is a
Figure 4. Specificity of phospho-TrkA antibodies. Affinity purification of polyclonal anti-
bodies raised against a short peptide in the C-terminal region of rat TrkA containing tyrosine
794,whichisphosphorylated in response to NGF (Loeb etal.,1994),was performed as described
in Materials and Methods. To assess specificity of purified antibodies, lysates (40
g each well)
obtained from PC12 cells treated with 50 ng/ml NGF for 10 min were probed with affinity-
purified serum as well as purified sera that was preincubated with either a phosphorylated or
nonphosphorylated TrkA peptide competitor.
6654 J. Neurosci., July 28, 2004 24(30):6650 6658 Rajagopal et al. Activation of Trk Receptors
potent inhibitor of Golgi vesicle fusion (Pelham, 1991). PC12
cells were treated with either 5
g/ml brefeldin A or vehicle for 3.5
hr. During this period of time, cells were simultaneously treated
with 10 nMCGS 21680 or 100 ng/ml NGF, such that the treatment
period would coincide with the end of brefeldin A treatment.
Untreated cells were used for control. Cell lysates were prepared
and used for Trk immunoprecipitation and subsequent analysis
by SDS-PAGE and blotting with phosphotyrosine antibodies. As
shown in Figure 6 (top panel), brefeldin A pretreatment com-
pletely abolished activation of Trk by CGS 21680 at both time
points of treatment, whereas NGF-mediated Trk activation was
left unimpaired.
In addition, Akt activation was also blocked when cells were
pretreated with brefeldin, indicating that activation and down-
stream signaling of Trk mediated by GPCRs requires an intact
Golgi apparatus. Interestingly, TrkA glycosylation was impaired
by brefeldin as indicated by the pan-Trk blot shown in Figure 6 A.
The 110 kDa isoform of Trk accumulated when cells were treated
with brefeldin. These results imply that an intact Golgi apparatus
is a potential location for Trk transactivation and that the positive
phospho-Trk staining observed in cells (Fig. 3A) may have arisen
from this organelle.
To determine how brefeldin A affected TrkA subcellular local-
ization, we subjected PC12 cells to indirect immunofluorescence
for TrkA and pTrkA after treatment with brefeldin A. As depicted
in Figure 6B, brefeldin A abolished perinuclear TrkA immuno-
reactivity in cells treated with CGS 21680 but did not affect the
ability of NGF to activate TrkA at the plasma membrane. This
indicated that Trk localization at the plasma membrane was not
greatly disrupted by brefeldin A.
Effect of transactivation on primary neurons
PACAP displays pronounced effects on the survival of septal cho-
linergic neurons (Takei et al., 2000) as well as many other neuro-
nal cells, such as mesencephalic dopaminergic neurons (Takei et
al., 1998) and sensory neurons (Lioudyno et al., 1998). PACAP
also is capable of triggering transactivation of Trk receptors in
primary neurons in a time course identical to adenosine or CGS
21680 treatment (Lee et al., 2002b). To determine the distribu-
tion of transactivated TrkA receptors in primary neurons, we
established E18 basal forebrain cultures. It was established previ-
ously that TrkA receptors are expressed by a subset of these neu-
rons (Bernd et al., 1988). After treatment with PACAP, the local-
ization of Trk receptors and phospho-Trk receptors was followed
using pan-Trk antibodies and the phospho-Trk-specific anti-
body. Similar to what was observed in cultured PC12 cells, the
phosphorylated form of Trk resided predominantly in an intra-
cellular location in basal forebrain neurons, whereas Trk recep-
tors could be found throughout the cell body and processes (Fig.
7). The phospho-Trk staining was heavily concentrated in a pe-
rinuclear region of the cell body, as illustrated by the DAPI and
phospho-Trk distribution. This pattern of staining was inhibited
by both K-252a and brefeldin A (Fig. 7), confirming that the
observed immunoreactivity represents activated TrkA in a Golgi
compartment. The finding of activated Trk receptors in response
to PACAP treatment in basal forebrain neurons confirms that
transactivation occurs in an intracellular localization in primary
neurons.
Discussion
Cross talk between GPCR ligands and receptor tyrosine kinases
has been described as a means of conveying proliferative signals
in cells (Downward, 2003; Fischer et al., 2003). In contrast, trans-
activation of neurotrophic receptors can result in survival re-
sponses and neuronal differentiation. In this study, we have
shown that Trk receptors undergo activation in intracellular
membranes and not at the cell surface. Biotinylation and immu-
nofluorescence measurements indicate that transactivation
events displayed a delayed time course and absence of involve-
Figure 5. Localization of transactivated TrkA receptors. Subcellular localization of transac-
tivated TrkA in PC12 cells was examined by confocal microscopy. A, Cells were treated with 10
nMCGS 21680 for 3 hr or with 100 ng/ml NGF for 5 min and then double labeled for phospho-
TrkA (rabbit polyclonal pTrkA; FITC-labeled goat anti-rabbit) and pan-Trk (monoclonal Trk B3;
Cy3-goat anti-mouse). B, To further characterize the specific location of transactivation events,
double labeling of CGS 21680-treated cells was performed with antibodies directed against
EEA-1, GM130, or TGN-38 (red) and phospho-TrkA (green).
Rajagopal et al. Activation of Trk Receptors J. Neurosci., July 28, 2004 24(30):6650 6658 6655
ment of cell surface receptors. These features are in direct con-
trast to activation of mitogenic growth factor receptors (Daub et
al., 1997; Luttrell et al., 1999; Prenzel et al., 1999).
The location of signaling events representing cross talk be-
tween GPCR ligands and Trk receptor tyrosine kinases plays a
crucial role in the kinetics and mechanism of transactivation.
Although EGF receptor transactivation by GPCR ligands is rapid
and requires endocytosis and close proximity of EGF receptor
and GPCRs (Maudsley et al., 2000; Pierce et al., 2001), the time
course of Trk receptor transactivation is longer and involves in-
tracellular processing events. For example, processing of imma-
ture forms of Trk is involved in transactivation.
Several other aspects of GPCR-mediated transactivation of
Trk receptors distinguish it from other transactivation and neu-
rotrophin signaling events. A noticeable difference between
adenosine and NGF Trk is that the phosphorylation of Trk sub-
strates, such as the Shc adaptor proteins PLC-
and PI3 kinase,
requires an extended period of time, 1 hr of adenosine treat-
ment (Lee and Chao, 2001), in contrast to NGF treatment, which
requires 1 min. Second, GPCR-mediated activation of Trk re-
sults in downstream signaling to phosphoinositide 3-kinase and
Akt but not MAPK, unlike neurotrophin signaling, which can
robustly activate both pathways. The specific activation of Akt by
transactivated Trk receptors provides a survival response in neu-
rons (Lee et al., 2001, 2002a). MAPK activation is the hallmark of
Figure 6. The Golgi apparatus is involved in Trk transactivation. A, PC12 cells were pre-
treated for 30 min with 5
g/ml brefeldin A or vehicle and then treated with CGS 21680 or NGF
as indicated. In all cases of brefeldin A treatment, cells were exposed to the drug for a total
duration of 3.5 hr. Trk immunoprecipitates were then analyzed by blotting with phosphoty-
rosine or pan-Trk antibodies. Phosphorylation of Akt was examined by blotting cell lysates with
either phospho-Akt or anti-Akt antibodies for activated and total Akt levels. B, PC12 cells pre-
treated with brefeldin A or vehicle were treated with 10 nMCGS 21680 for 3 hr or 100 ng/ml NGF
for 5 min and then stained for pTrkA (green) or total Trk (red) and imaged by standard immu-
nofluorescent microscopy. Cell nuclei were visualized with DAPI.
Figure 7. Localization of transactivated TrkA in basal forebrain neurons. Indirect immuno-
fluorescence analysis was used to examine localization of transactivated TrkA in primary basal
forebrain neuronal cultures. Cultures were treated with PACAP38 as indicated and labeled with
phospho-TrkA (green) and pan-Trk (red) antibodies. Where indicated, K-252a (100 nM)or
brefeldin A (5
g/ml) was used to pretreat cells before addition of PACAP38. Cell nuclei were
stained with DAPI (blue).
6656 J. Neurosci., July 28, 2004 24(30):6650 6658 Rajagopal et al. Activation of Trk Receptors
many growth factor receptors, such as EGF and PDGF receptors,
that provide proliferative signals after transactivation (Luttrell et
al., 1999; Wetzker and Bohmer, 2003).
Transactivation by adenosine or PACAP does not involve the
production or release of newly synthesized or processed neuro-
trophins, as has been observed for the EGF receptor (Prenzel et
al., 1999). One mechanism for transactivation of the EGF recep-
tor involves the action of matrix metalloproteases. Cleavage of
membrane-bound proHB-EGF by metalloproteases results in the
release of EGF that binds and activates the EGF receptor (Prenzel
et al., 1999). Experiments performed in PC12 cells indicate that
GPCR ligands did not result in an increase in the expression of
NGF that might contribute to TrkA receptor activation (Fig. 1C)
(Lee et al., 2002a). Also, binding experiments show that adeno-
sine and PACAP do not displace NGF from binding to TrkA
receptors, indicating that the effects of these GPCR ligands occur
through an intracellular pathway.
What is the relevance of these transactivation events? During
retrograde transport, complexes of NGF with TrkA in clathrin-
coated vesicles and endosomes have been envisioned, giving rise
to the model of NGF and TrkA as components of a retrograde
signal (Riccio et al., 1997; Senger and Campenot, 1997; Ginty and
Segal, 2002; Grimes and Miettinen, 2003). A number of signaling
molecules have been found to be associated with the TrkA recep-
tor during retrograde transport (Delcroix et al., 2003). This sug-
gests that signaling by trophic factors persists after internalization
of their receptors in intracellular locations. Implicit in this model
is that internalization of NGF is necessary for eliciting survival
signals at the cell body. The evidence comes from the activation of
the CREB transcription factor (Riccio et al., 1997) and Erk5
(Watson et al., 2001) by retrograde signals at distal terminals.
Retrograde signaling events, however, may also be transmit-
ted in the absence of NGF internalization and transport. Survival
of sympathetic neurons can be mediated by transported Trk re-
ceptors in the cell body after stimulation at nerve terminals by
NGF linked to beads (MacInnis and Campenot, 2002). This raises
the possibility that mechanisms other than endocytosed NGF
may propagate signals from distal axons to TrkA receptors at the
cell body (Campenot and MacInnis, 2004). A number of argu-
ments have been raised both for and against the hypothesis that
retrograde signaling can occur in the absence of NGF uptake
(Chao 2003b; Ye et al., 2003), and several diverse models have
been proposed to account for the discrepancies in retrograde
signaling mechanisms (Ginty and Segal, 2002; Heerssen and Se-
gal, 2002; Miller and Kaplan, 2003).
One common feature of retrograde transport of neurotrophin
signaling is the ability of internalized Trk receptors to signal in a
productive manner. Activation of tyrosine kinase receptors from
GPCR transactivation illustrates an alternative way in which sig-
nals may be transmitted by intracellular receptors in the absence
of ligand. This is exemplified by the findings in this study in
which adenosine is capable of activating Trk receptors in intra-
cellular membranes and not at the cell surface. Activation of in-
tracellular Trk receptors may depend on Src kinase activity, re-
cruitment of adaptor proteins, or an increase in the local
concentration of Trk, which could lead to constitutive activation.
The results demonstrate that internal Trk receptors may be found
in discrete organelles (Grimes et al., 1997; Jullien et al., 2002;
Delcroix et al., 2003), where they are fully active and promote Akt
survival signals (Lee et al. 2001, 2002a,b). Also, it has been estab-
lished that TrkA receptors found in sympathetic neurons may be
activated by engagement of the Ret tyrosine kinase by GDNF in
the absence of NGF (Tsui-Pierchala et al., 2002). Hence, these
observations solidify the growing realization that Trk receptors
function not only at the cell membrane but provide potent signals
from discrete intracellular locations without neurotrophin bind-
ing. Trafficking of neurotrophin receptors in different mem-
brane compartments must play a decisive role in regulating var-
ious fundamental signaling pathways in neurons.
References
Aibel L, Martin-Zanca D, Perez P, Chao M (1998) Functional expression of
TrkA receptors in hippocampal neurons. J Neurosci Res 54:424431.
Bernd P, Martinez H, Dreyfus C, Black I (1988) Localization of high-affinity
and low-affinity nerve growth factor receptors in cultured rat basal fore-
brain. Neuroscience 26:121129.
Bronfman F, Tcherpakov M, Jovin T, Fainzilber M (2003) Ligand-induced
internalization of the p75 neurotrophin receptor: a slow route to the
signaling endosome. J Neurosci 23:32093220.
Campenot RB, MacInnis BL (2004) Retrograde transport of neurotrophins:
fact and function. J Neurobiol 58:217229.
Chao MV (2003a) Neurotrophins and their receptors: a convergence point
for many signaling pathways. Nat Rev Neurosci 4:299309.
Chao MV (2003b) Retrograde transport redux. Neuron 39:12.
Daub H, Weiss FU, Wallasch C, Ullrich A (1996) Role of transactivation of
the EGF receptor in signaling by G-protein coupled receptors. Nature
379:557560.
Daub H, Wallash C, Lankenau A, Herrlich A, Ullrich A (1997) Signal char-
acteristics of G protein-transactivated EGF receptor. EMBO J
16:70327044.
Delcroix J-D, Valetta J, Wu C, Hunt S, Kowal A, Mobley W (2003) NGF
signaling in sensory neurons: evidence that early endosomes carry NGF
retrograde signals. Neuron 39:6984.
DiStefano PS, Friedman B, Radziejewski C, Alexander C, Boland P, Schick
CM, Lindsay RM, Wiegand SJ (1992) The neurotrophins BDNF, NT-3,
and NGF display distinct patterns of retrograde axonal-transport in pe-
ripheral and central neurons. Neuron 8:983993.
Donovan MJ, Miranda RC, Kraemer R, McCaffrey TA, Tessarollo L, Ma-
hadeo D, Sharif S, Kaplan DR, Tsoulfas P, Parada L, Toran-Allerand CD,
Hajjar DP, Hempstead BL (1995) Neurotrophin and neurotrophin re-
ceptors in vascular smooth muscle cells. Regulation of expression in re-
sponse to injury. Am J Pathol 147:309324.
Downward J (2003) Role of receptor tyrosine kinases in G-protein-coupled
receptor regulation of Ras: transactivation or parallel pathways. Biochem
J 376:e9e10.
Fischer OM, Hart S, Gschwind A, Ullrich A (2003) EGFR signal transacti-
vation in cancer cells. Biochem Soc Trans 31:12031208.
Ginty D, Segal R (2002) Retrograde neurotrophin signaling: Trk-ing along
the axon. Curr Opin Neurobiol 12:268274.
Grimes M, Miettinen H (2003) Receptor tyrosine kinase and G-protein
coupled receptor signaling and sorting within endosomes. J Neurochem
84:905918.
Grimes M, Beattie E, Mobley W (1997) A signaling organelle containing the
nerve growth factor-activated receptor tyrosine kinase, TrkA. Proc Natl
Acad Sci USA 94:99099914.
Heerssen HM, Segal RA (2002) Location, location, location: a spatial view of
neurotrophin signal transduction. Trends Neurosci 25:160165.
Hempstead B (2002) The many faces of p75NTR. Curr Opin Neurobiol
12:260267.
Hempstead BL, Rabin SJ, Kaplan L, Reid S, Parada LF, Kaplan DR (1992)
Overexpression of the Trk tyrosine kinase rapidly accelerates nerve
growth factor-induced differentiation. Neuron 9:883896.
Hendry I, Stoeckel K, Thoenen H, Iversen L (1974) The retrograde axonal
transport of nerve growth factor. Brain Res 68:103121.
Huang E, Reichardt L (2001) Neurotrophins: roles in neuronal develop-
ment and function. Annu Rev Neurosci 24:677736.
Huang E, Reichardt L (2003) Trk receptors: roles in neuronal signal trans-
duction. Annu Rev Biochem 72:609642.
Ishihara T, Shigemoto R, Mori K, Takahashi K, Nagata S (1992) Functional
expression and tissue distribution of a novel receptor for vasoactive intes-
tinal polypeptide. Neuron 8:811819.
Jarvis M, Schulz R, Hutchinson A, Do U, Sills M, Williams M (1989)
[
3
H]CGS21680, a selective A2 adenosine receptor agonist directly labels
A2 receptors in rat brain. J Pharmacol Exp Ther 251:888893.
Jullien J, Guilli V, Reichardt L, Rudkin B (2002) Molecular kinetics of nerve
Rajagopal et al. Activation of Trk Receptors J. Neurosci., July 28, 2004 24(30):6650 6658 6657
growth factor receptor trafficking and activation. J Biol Chem
277:3870038708.
Kim K, von Zastrow M (2003) Neurotrophin-regulated sorting of opioid
receptors in the biosynthetic pathway of neurosecretory cells. J Neurosci
23:20752085.
Kotecha S, Oak J, Jackson M, Perez Y, Orser B, Van Tol H, MacDonald J
(2002) A D2 class dopamine receptor transactivates a receptor tyrosine
kinase to inhibit NMDA receptor transmission. Neuron 35:11111122.
Lee F, Rajagopal R, Chao M (2002a) Distinctive features of Trk neurotro-
phin receptor transactivation by G protein-coupled receptors. Cytokine
Growth Factor Rev 13:1117.
Lee F, Rajagopal R, Kim A, Chang P, Chao M (2002b) Activation of Trk
neurotrophin receptor signaling by pituitary adenylate cyclase-activating
polypeptides. J Biol Chem 277:90969102.
Lee FS, Chao MV (2001) Activation of Trk neurotrophin receptors in the
absence of neurotrophins. Proc Natl Acad Sci USA 92:35553560.
Lioudyno M, Skoglosa Y, Takei N, Lindholm D (1998) Pituitary adenylate
cyclase-activating polypeptide (PACAP) protects dorsal root ganglion
neurons from death and induces calcitonin gene-related peptide (CGRP)
immunoreactivity in vitro. J Neurosci Res 51:243256.
Loeb DM, Stephens RM, Copeland T, Kaplan DR, Greene LA (1994) A Trk
nerve growth-factor (NGF) receptor point mutation affecting interaction
with phospholipase c-gamma-1 abolishes NGF-promoted peripherin in-
duction but not neurite outgrowth. J Biol Chem 269:89018910.
Luttrell L, Daaka Y, Lefkowitz R (1999) Regulation of tyrosine kinase cas-
cades by G-protein-coupled receptors. Curr Opin Cell Biol 11:177183.
Luzio J, Brake B, Banting G, Howell K, Braghetta P, Stanley K (1990) Iden-
tification, sequencing and expression of an integral membrane protein of
the trans-Golgi network (TGN38). Biochem J 270:97102.
MacInnis B, Campenot R (2002) Retrograde support of neuronal survival
without retrograde transport of nerve growth factor. Science
295:15361539.
Maudsley S, Pierce K, Musa Zamah A, Miller W, Ahn S, Daaka Y, Lefkowitz R,
Luttrell L (2000) The
2-adrenergic receptor mediates extracellular
signal-regulated kinase activation via assembly of a multi-receptor com-
plex with the epidermal growth factor receptor. J Biol Chem
275:95729580.
Miller FD, Kaplan DR (2003) Neurobiology. Trk makes the retrograde. Sci-
ence 295:14711473.
Nakamura N, Rabouille C, Watson R, Nilsson T, Hui N, Slusarewicz P, Kreis
T, Warren G (1995) Characterization of a cis-Golgi matrix protein,
GM130. J Cell Biol 131:17151726.
Pelham H (1991) Multiple targets for brefeldin A. Cell 67:449451.
Peng Y, Amemiya M, Yang X, Fan L, Moe O, Yin H, Preisig P, Yanagisawa M,
Alpern R (2001) ETB receptor activation causes exocytic insertion of
NHE3 in OKP cells. Am J Physiol Renal Physiol 280:F34F42.
Pierce K, Tohgo A, Ahn S, Field M, Luttrell L, Lefkowitz R (2001) Epidermal
growth factor (EGF) receptor-dependent ERK activation by G protein-
coupled receptors. J Biol Chem 276:2315523160.
Prenzel N, Zwick E, Daub H, Leserer M, Abraham R, Wallasch C, Ullrich A
(1999) EGF receptor transactivation by G-protein-coupled receptors re-
quires metalloproteinase cleavage of proHB-EGF. Nature 402:884888.
Riccio A, Pierchala B, Ciarallo C, Ginty D (1997) An NGF-TrkA-mediated
retrograde signal to transcription factor CREB in sympathetic neurons.
Science 277:10971100.
Senger D, Campenot R (1997) Rapid retrograde tyrosine phosphorylation
of TrkA and other proteins in rat sympathetic neurons in compartmented
cultures. J Cell Biol 138:411421.
Takei N, Skoglosa Y, Lindholm D (1998) Neurotrophic and neuroprotec-
tive effects of pituitary adenylate cyclase-activating polypeptide (PACAP)
on mesencephalic dopaminergic neurons. J Neurosci Res 54:698706.
Takei N, Torres E, Yuhara A, Jongsma H, Otto C, Korhonen L, Abiru Y,
Skoglosa Y, Schultz G, Hatanaka H, Sofroniew M, Lindholm D (2000)
Pituitary adenylate cyclase-activating polypeptide promotes the survival
of basal forebrain cholinergic neurons in vitro and in vivo: comparison
with effects of nerve growth factor. Eur J Neurosci 12:22732280.
Tsui-Pierchala BA, Milbrandt J, Johnson E (2002) NGF utilizes c-Ret via a
novel GFL-independent, inter-RTK signaling mechanism to maintain the
trophic status of mature sympathetic neurons. Neuron 33:261273.
Tsui-Pierchala BA, Ginty DD (1999) Characterization of an NGF-P-TrkA
retrograde-signaling complex and age-dependent regulation of TukA
phosphorylation in sympathetic neurons. J Neurosci 19:82078218.
Watson F, Porcionatto M, Bhattacharyya A, Stiles C, Segal R (1999a) Trk
glycosylation regulates receptor localization and activity. J Neurobiol
39:323336.
Watson F, Heerssen H, Moheban D, Lin M, Sauvageot C, Bhattacharyya A,
Pomeroy S, Segal R (1999b) Rapid nuclear responses to target-derived
neurotrophins require retrograde transport of ligand-receptor complex.
J Neurosci 19:78897900.
Watson F, Heerssen H, Bhattacharyya A, Klesse L, Lin M, Segal R (2001)
Neurotrophins use the Erk5 pathway to mediate a retrograde survival
response. Nat Neurosci 4:981988.
Wetzker R, Bohmer FD (2003) Transactivation joins multiple tracks to the
ERK/MAPK cascade. Nat Rev Mol Cell Biol 4:651657.
Ye H, Kuruvilla R, Zeifel L, Ginty D (2003) Evidence in support of signaling
endosome-based retrograde survival of sympathetic neurons. Neuron
39:5768.
York R, Yao H, Dillon T, Ellig C, Eckert S, McCleskey E, Stork P (1998) Rap1
mediates sustained MAP kinase activation induced by nerve growth fac-
tor. Nature 392:622626.
6658 J. Neurosci., July 28, 2004 24(30):6650 6658 Rajagopal et al. Activation of Trk Receptors
... Considering the transactivation crosstalk between GPCRs and receptor tyrosine kinases, including TrkB (Rajagopal et al., 2004;2006;El Zein et al., 2007), and recent evidence for GPCR modulation/self-association by flavonoids on the latter (Herrera-Hernández et al., 2017; Ortega et al., 2019), we speculated that one mechanism by which cannflavins could interfere on Arc mRNA expression in cortical neurons involves activation of a G protein signal that transinactivates the function of molecular cascades downstream of TrkB receptors responsible for Arc expression. To explore this scenario, we tested the effect of cannflavins on the GPCRome, en masse, using the PRESTO-Tango assay-an unbiased high-throughput screening approach adapted to identify agonist activity of agents towards the large family of GPCRs (Kroeze et al., 2015). ...
... Nevertheless, two specific results from our study support to a certain degree the idea that a direct functional interaction most likely occur between cannflavins and TrkB. First, our interrogation of the GPCRome using the PRESTO-Tango assay showed that cannflavins do not stimulate the activity of more than 300 GPCRs, which rules out the possibility that cannflavins are disrupting TrkB function through a GPCR transinactivation event in our cellular experiments (Rajagopal et al., 2004;2006;El Zein et al., 2007). And second, our experiment with Neuro2a cells stably expressing TrkB reveals that cannflavins block the growth of neurites, as well as the observed cell survival stimulatory effect, produced by exogenous BDNF application in this model. ...
Article
Full-text available
Cannflavins A and B are flavonoids that accumulate in the Cannabis sativa plant. These specialized metabolites are uniquely prenylated and highly lipophilic, which may permit their interaction with membrane-bound enzymes and receptors. Although previous studies found that cannflavins can produce anti-inflammatory responses by inhibiting the biosynthesis of pro-inflammatory mediators, the full extent of their cellular influence remains to be understood. Here, we studied these flavonoids in relation to the Tropomyosin receptor kinase B (TrkB), a receptor tyrosine kinase that is activated by the growth factor brain-derived neurotrophic factor (BDNF). Using mouse primary cortical neurons, we first collected evidence that cannflavins prevent the accumulation of Activity-regulated cytoskeleton-associated (Arc) protein upon TrkB stimulation by exogenous BDNF in these cells. Consistent with this effect, we also observed a reduced activation of TrkB and downstream signaling effectors that mediate Arc mRNA transcription when BDNF was co-applied with the cannflavins. Of note, we also performed a high-throughput screen that demonstrated a lack of agonist action of cannflavins towards 320 different G protein-coupled receptors, a result that specifically limit the possibility of a TrkB transinactivation scenario via G protein signaling to explain our results with dissociated neurons. Finally, we used Neuro2a cells overexpressing TrkB to show that cannflavins can block the growth of neurites and increased survival rate produced by the higher abundance of the receptor in this model. Taken together, our study offers a new path to understand the reported effects of cannflavins and other closely related compounds in different cellular contexts.
... Phase 2 basket trial of taletrectinib for solid tumors with NTRK initiated TRK receptors can be activated by different mechanisms, such as the G-protein coupled receptor (GPCR) mechanism, without neurotrophins' involvement. In order to increase the survival of neural cells through AKT activity, the TRK receptor can be activated by two GPCR ligands, adenosine, and pituitary adenylate cyclase-activating polypeptide (PACAP) [20]. Somatic NTRK mutation has been identified in various tumor types, including colorectal and lung cancers such as large cell neuroendocrine carcinoma and NSCLC, as well as melanoma and acute myeloid leukemia [21][22][23][24]. ...
Article
Full-text available
Neurotrophic tyrosine receptor kinase (NTRK) has been a remarkable therapeutic target for treating different malignancies, playing an essential role in oncogenic signaling pathways. Groundbreaking trials like NAVIGATE led to the approval of NTRK inhibitors by the Food and Drug Administration (FDA) to treat different malignancies, significantly impacting current oncology treatment. Accurate detection of NTRK gene fusion becomes very important for possible targeted therapy. Various methods to detect NTRK gene fusion have been applied widely based on sensitivity , specificity, and accessibility. The utility of different tests in clinical practice is discussed in this study by providing insights into their effectiveness in targeting patients who may benefit from therapy. Widespread use of NTRK inhibitors in different malignancies could remain limited due to resistance mechanisms that cause challenges to medication efficacy in addition to common side effects of the medications. This review provides a succinct overview of the application of NTRK inhibitors in various types of cancer by emphasizing the critical clinical significance of NTRK fusion gene detection. The discussion also provides a solid foundation for understanding the current challenges and potential changes for improving the efficacy of NTRK inhibitor therapy to treat different malig-nancies.
... Mounting evidence indicates that synergism between adenosine A 2A R and BDNF receptors is crucial for a suitable neuronal function and synaptic transmission. This includes the maintenance of adequate levels of BDNF and TrkB-FL receptors and consequently its mediated actions, such as the preservation of long-term potentiation (LTP), considered an electrophysiological model for the basic mechanisms involved in learning and memory formation [19][20][21][22][23][24][25][26][27]. Impairments in the A 2A R/BDNF crosstalk were observed in symptomatic Mecp2 −/y mice. ...
Article
Full-text available
Rett Syndrome is an X-linked neurodevelopmental disorder (RTT; OMIM#312750) associated to MECP2 mutations. MeCP2 dysfunction is seen as one cause for the deficiencies found in brain-derived neurotrophic factor (BDNF) signaling, since BDNF is one of the genes under MeCP2 jurisdiction. BDNF signaling is also dependent on the proper function of the adenosinergic system. Indeed, both BDNF signaling and the adenosinergic system are altered in Mecp2-null mice (Mecp2−/y), a representative model of severe manifestation of RTT. Considering that symptoms severity largely differs among RTT patients, we set out to investigate the BDNF and ADO signaling modifications in Mecp2 heterozygous female mice (Mecp2+/−) presenting a less severe phenotype. Symptomatic Mecp2+/− mice have lower BDNF levels in the cortex and hippocampus. This is accompanied by a loss of BDNF-induced facilitation of hippocampal long-term potentiation (LTP), which could be restored upon selective activation of adenosine A2A receptors (A2AR). While no differences were observed in the amount of adenosine in the cortex and hippocampus of Mecp2+/− mice compared with healthy littermates, the density of the A1R and A2AR subtype receptors was, respectively, upregulated and downregulated in the hippocampus. Data suggest that significant changes in BDNF and adenosine signaling pathways are present in an RTT model with a milder disease phenotype: Mecp2+/− female animals. These features strengthen the theory that boosting adenosinergic activity may be a valid therapeutic strategy for RTT patients, regardless of their genetic penetrance.
... Together, these studies further demonstrate the potential utility of TrkB ligands in replacing or mimicking the action of BDNF. In addition to ligand-induced effects, TrkB receptors can also be transactivated by other receptor systems such as the G-protein-coupled receptors (GPCRs) (Rajagopal et al., 2004) ( Figure 2). Transactivation elicited by these GPCR ligands leads to neuroprotective effects (Takei et al., 2000), providing an alternative mechanism for modulating BDNF signaling in the CNS. ...
Article
Full-text available
Being the highest expressed neurotrophin in the mammalian brain, the brain‐derived neurotrophic factor (BDNF) is essential to neural development and plasticity in both health and diseases. Following the discovery of BDNF by Yves‐Alain Barde in 1982, the main feature of BDNF's activity in myelination was first described by Cellerino et al. in 1997. Since then, genetic manipulation of the BDNF‐encoding gene and its receptors in murine models has revealed the contribution of BDNF to the myelinating process in the central nervous system (CNS). The series of BDNF or receptor mouse mutants as well as the BDNF polymorphism in humans have provided new insights into the roles that BDNF signaling plays in myelination in a complex manner. 2024 marks the 30th year of BDNF's research in myelination. Here, we share our perspective on the 30‐year history of BDNF in the field of CNS myelination from phenotyping to therapeutic development, focusing on genetic evidence regarding the mechanism by which BDNF regulates myelin formation and repair in the CNS. This review also discusses the current hypotheses of BDNF's action on CNS myelination: axonal‐ and oligodendroglial‐driven mechanisms, which may be ultimately activity‐dependent. Last, this review raises the challenges and opportunities of developing BDNF‐based therapies for neurodegenerative diseases, opening unanswered questions for future investigation. image
... The binding of neurotrophins induces the activation of the EPAC downstream signaling molecule ERK1/2, leading to CREB phosphorylation and growth cone preservation in neurons [28]. Additionally, GPCRs, e.g., adenosine A 2A receptor or procaspase activating compound-1 (PAC1) receptor activation, modulate neuroregeneration through AC-mediated cAMP production, leading to transactivation of the Trk receptors in the absence of neurotrophins [29]. Furthermore, the Gα s unit is crucial to increasing Trk receptor translocation to the cellular surface and hence enhancing Trk signaling [30]. ...
Article
Full-text available
Traumatic spinal cord injury (SCI) is characterized by severe neuroinflammation and hampered neuroregeneration, which often leads to permanent neurological deficits. Current therapies include decompression surgery, rehabilitation, and in some instances, the use of corticosteroids. However, the golden standard of corticosteroids still achieves minimal improvements in functional outcomes. Therefore, new strategies tackling the initial inflammatory reactions and stimulating endogenous repair in later stages are crucial to achieving functional repair in SCI patients. Cyclic adenosine monophosphate (cAMP) is an important second messenger in the central nervous system (CNS) that modulates these processes. A sustained drop in cAMP levels is observed during SCI, and elevating cAMP is associated with improved functional outcomes in experimental models. cAMP is regulated in a spatiotemporal manner by its hydrolyzing enzyme phosphodiesterase (PDE). Growing evidence suggests that inhibition of cAMP-specific PDEs (PDE4, PDE7, and PDE8) is an important strategy to orchestrate neuroinflammation and regeneration in the CNS. Therefore, this review focuses on the current evidence related to the immunomodulatory and neuroregenerative role of cAMP-specific PDE inhibition in the SCI pathophysiology.
... In contrast to the BDNF-induced receptor activation, transactivation of TrkB via adenosine appears slower and seems to selectively activate the PI3K/Akt signaling pathway to promote cell survival (Lee and Chao, 2001;Lee et al., 2002a;Wiese et al., 2007). Furthermore, G protein-coupled receptors (GPCR), activated by pituitary adenylate cyclaseactivating polypeptide, or dopamine, could effectively trigger TrkB transactivation and its surface expression (Iwakura et al., 2008;Lee et al., 2002b;Rajagopal et al., 2004). Similarly, the epidermal growth factor (EGF), via its receptor EGFR, is able to induce TrkB activation in cortical precursor cells to control their migration within the cortex (Puehringer et al., 2013). ...
Thesis
Full-text available
Spinal muscular atrophy (SMA) is a genetic pediatric condition that affects lower motoneurons leading to their degeneration and muscle weakness. It is caused by homozygous loss or mutations in the Survival Motor Neuron 1 (SMN1) gene; however, the pathomechanism leading to motoneuron degeneration is not fully resolved. Cultured embryonic SMA motoneurons display axon elongation and differentiation defects accompanied by collapsed growth cones with a disturbed actin cytoskeleton. Intriguingly, motoneurons cultured from mice deficient for the Tropomyosin-kinase receptor B (TrkB), exhibit similar pathological features. Thus, the question arises whether SMA motoneurons suffer from defective Brain-derived neurotrophic factor (BDNF)/TrkB signaling and whether there is a link to the disturbed actin cytoskeleton. In the recent years, modifier genes such as Plastin 3 (PLS3) were shown to beneficially interfere with SMA pathology. Nevertheless, the mechanism of how the actin-bundler PLS3 counteracts SMN deficiency is not well understood. In this study, we investigated TrkB localization and its activation in cultured SMA motoneurons and neuromuscular junctions (NMJs). While TrkB levels are only mildly affected locally in axon terminals, BDNF-mediated TrkB phosphorylation was massively disturbed. The activity-dependent TrkB translocation to the cell surface and its activation via BDNF were shown to be Pls3-dependent processes, that can be abolished by knockdown of Pls3. In contrast, PLS3 overexpression in SMA motoneurons rescued the defects on morphological and functional level. In particular, the relocation of TrkB to the cell surface after BDNF-induced internalization is disturbed in SMA, which is based on an actin-dependent TrkB translocation defect from intracellular stores. Lastly, AAV9-mediated PLS3 overexpression in vivo in neonatal SMA mice provided further evidence for the capacity of PLS3 to modulate actin dynamics necessary for accurate BDNF/TrkB signaling. In conclusion, we provide a novel role for PLS3 in mediating proper alignment of transmembrane proteins as prerequisite for their appropriate functioning. Hence, PLS3 is required for a key process indispensable for the development and function of motoneurons even beyond the context of SMA.
Article
Transactivation of Tropomyosin receptor kinase B (TrkB) by EGF leads to cell surface transport of TrkB, promoting its signaling responsiveness to brain-derived neurotrophic factor (BDNF), a critical process for proper cortical plate development. However, the mechanisms that regulate the transport of TrkB to the cell surface are not fully understood. Here, we identified Calnexin as a regulator for targeting TrkB either to the cell surface or toward autophagosomal processing. Calnexin-deficient mouse embryos show impaired cortical plate formation and elevated levels of transactivated TrkB. In Calnexin-depleted mouse neuronal precursor cells, we detected an impaired cell surface transport of TrkB in response to EGF and an impaired delivery to autophagosomes. Mechanistically, we show that Calnexin facilitates the interaction of TrkB with the ER-phagy receptor Fam134b, thereby targeting TrkB to ER-phagy. This mechanism appears as a critical process for fine-tuning the sensitivity of neurons to BDNF.
Chapter
The Oxford Handbook of the Neurobiology of Pain represents a state of the art overview of the rapidly developing field of pain research. As populations age, the number of people in pain is growing dramatically, with half the population living with pain. The opioid crisis has highlighted this problem. The present volume is thus very timely, providing expert overviews of many complex topics in pain research that are likely to be of interest not just to pain researchers, but also to pain clinicians who are seeking new therapeutic opportunities to develop analgesics. Many of the topics covered are of interest to neuroscientists, as pain is one of the most amenable sensations for mechanistic dissection. The present volume covers all aspects of the topic, from a history of pain through invertebrate model systems to the human genetics of pain and functional imaging. Chapters include the role of ion channels, the opioid system, the immune and sympathetic systems, as well as the mechanisms that transform acute to chronic pain. Migraine and the interplay between sleep and pain are also discussed. New technology in the form of transgenic animals, chemogenetics, optogenetics, and proteomic analyses are providing significant advances in our research and are covered as well. Demystifying pain through an understanding of its fundamental biology, as outlined in this volume, is the most direct route to ameliorating this vast human problem.
Article
Antisera raised to a detergent- and salt-resistant matrix fraction from rat liver Golgi stacks were used to screen an expression library from rat liver cDNA. A full-length clone was obtained encoding a protein of 130 kD (termed GM130), the COOH-terminal domain of which was highly homologous to a Golgi human auto-antigen, golgin-95 (Fritzler et al., 1993). Biochemical data showed that GM130 is a peripheral cytoplasmic protein that is tightly bound to Golgi membranes and part of a larger oligomeric complex. Predictions from the protein sequence suggest that GM130 is an extended rod-like protein with coiled-coil domains. Immunofluorescence microscopy showed partial overlap with medial- and trans-Golgi markers but almost complete overlap with the cis-Golgi network (CGN) marker, syntaxin5. Immunoelectron microscopy confirmed this location showing that most of the GM130 was located in the CGN and in one or two cisternae on the cis-side of the Golgi stack. GM130 was not re-distributed to the ER in the presence of brefeldin A but maintained its overlap with syntaxin5 and a partial overlap with the ER-Golgi intermediate compartment marker, p53. Together these results suggest that GM130 is part of a cis-Golgi matrix and has a role in maintaining cis-Golgi structure.
Article
Vasoactive intestinal polypeptide (VIP), a 28 amino acid peptide hormone, plays many physiological roles in the peripheral and central nerve systems. A functional cDNA clone of the VIP receptor was isolated from a rat lung cDNA library by cross-hybridization with the secretin receptor cDNA. VIP bound the cloned VIP receptor expressed in mouse COP cells and stimulated adenylate cyclase through the cloned receptor. The rat VIP receptor consists of 459 amino acids with a calculated M, of 52,054 and contains seven transmembrane segments. It is structurally related to the secretin, calci-tonin, and parathyroid hormone receptors, suggesting that they constitute a new subfamily of the G, protein-coupled receptors. VIP receptor mRNA was detected in various rat tissues including liver, lung, intestines, and brain. In situ hybridization revealed that VIP receptor mRNA is widely distributed in neuronal cells of the adult rat brain, with a relatively highexpression in thecerebral cortex and hippocampus.
Article
Vasoactive intestinal polypeptide (VIP), a 28 amino acid peptide hormone, plays many physiological roles in the peripheral and central nerve systems. A functional cDNA clone of the VIP receptor was isolated from a rat lung cDNA library by cross-hybridization with the secretin receptor cDNA. VIP bound the cloned VIP receptor expressed in mouse COP cells and stimulated adenylate cyclase through the cloned receptor. The rat VIP receptor consists of 459 amino acids with a calculated M, of 52,054 and contains seven transmembrane segments. It is structurally related to the secretin, calci-tonin, and parathyroid hormone receptors, suggesting that they constitute a new subfamily of the G, protein-coupled receptors. VIP receptor mRNA was detected in various rat tissues including liver, lung, intestines, and brain. In situ hybridization revealed that VIP receptor mRNA is widely distributed in neuronal cells of the adult rat brain, with a relatively highexpression in thecerebral cortex and hippocampus.
Article
In the present study, the binding of a highly A2-selective agonist radioligand, (3H)CGS 21680 (2-(p-(2-carboxyethyl)-phenethylamino)-5'-N-ethylcarboxamido adenosine) is described. (3H)CGS 21680 specific binding to rat striatal membranes was saturable, reversible and dependent upon protein concentration. Saturation studies revealed that (3H)CGS 21680 bound with high affinity (Kd = 15.5 nM) and limited capacity (apparent Bmax = 375 fmol/mg of protein) to a single class of recognition sites. Estimates of ligand affinity (16 nM) determined from association and dissociation kinetic experiments were in close agreement with the results from the saturation studies. (3H)CGS 21680 binding was greatest in striatal membranes with negligible specific binding obtained in rat cortical membranes. Adenosine agonists ligands competed for the binding of 5 nM (3H)CGS 21680 to striatal membranes with the following order of activity; CGS 21680 = 5'-N-ethylcarboxamidoadenosine greater than 2-phenylaminoadenosine (CV-1808) = 5'-N-methylcarboxamidoadenosine = 2-chloroadenosine greater than R-phenylisopropyladenosine greater than N6-cyclohexyladenosine greater than N6cyclopentyltheophylline greater than S-phenylisopropyladenosine. The nonxanthine adenosine antagonist, CGS 15943A, was the most active compound in inhibiting the binding of (3H)CGS 21680. Other adenosine antagonists inhibited binding in the following order; xanthine amine congener = 1,3-dipropyl-8-(2-amino-4-chloro)phenylxanthine greater than 1,3-dipropyl-8-cyclopentylxanthine greater than 1,3-diethyl-8-phenylxanthine greater than 8-phenyltheophylline greater than 8-cyclopentyltheophylline = xanthine carboxylic acid congener greater than 8-parasulfophenyltheophylline greater than theophylline greater than caffeine.
Article
The human nerve growth factor receptor (TrkA) contains four potential N-glycosylation sites that are highly conserved within the Trk family of neurotrophin receptors, and nine additional sites that are less well conserved. Using a microscale deglycosylation assay, we show here that both conserved and variable N-glycosylation sites are used during maturation of TrkA. Glycosylation at these sites serves two distinct functions. First, glycosylation is necessary to prevent ligand-independent activation of TrkA. Unglycosylated TrkA core protein is phosphorylated even in the absence of ligand stimulation and displays constitutive kinase activity as well as constitutive interaction with the signaling molecules Shc and PLC-γ. Second, glycosylation is required to localize TrkA to the cell surface, where it can trigger the Ras/Raf/MAP kinase cascade. Using confocal microscopy, we show that unglycosylated active Trk receptors are trapped intracellularly. Furthermore, the unglycosylated active TrkA receptors are unable to activate kinases in the Ras-MAP kinase pathway, MEK and Erk. Consistent with these biochemical observations, unglycosylated TrkA core protein does not promote neuronal differentiation in Trk PC12 cells even at high levels of constitutive catalytic activity. © 1999 John Wiley & Sons, Inc. J Neurobiol 39: 323–336, 1999
Article
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a member of the vasointestinal polypeptide gene family for which neurotrophic activity has been postulated. PACAP mRNA is expressed in the developing and adult hippocampus, which is the principal target region of septal cholinergic neurons. We therefore studied the effects of PACAP on septal cholinergic neurons. In primary cultures from septum of embryonic and postnatal rats, PACAP increased the number of neurons immunohistochemically stained for the low-affinity nerve growth factor (NGF) receptor p75 and for the enzyme choline acetyltransferase (ChAT). PACAP also caused a corresponding increase in ChAT activity. In comparison, NGF had a greater effect than PACAP on the number of p75- and ChAT-positive neurons in these cultures. In vivo, following fimbria fornix transection, the number of immunohistochemically stained septal cholinergic neurons fell significantly to 18% in rats given continuous intracerebroventricular infusion of vehicle, whereas in rats given NGF the number of these neurons did not differ significantly from unoperated controls. In PACAP-treated rats the number was 48% of unoperated values, which represented a significant increase compared with vehicle-treated rats and a significant decrease compared with NGF-treated rats or unoperated controls. Double-staining experiments revealed that most ChAT-positive neurons in rat medial septum also express PACAP receptor 1. Together the results show that PACAP promotes the survival of septal cholinergic neurons in vitro, and after injury in vivo, suggesting that PACAP acts as a neurotrophic factor influencing the development and maintenance of these neurons.
Article
The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is present in many regions of the adult and developing brain as are receptors for PACAP. PACAP stimulates different signalling cascades in neurons, involving cAMP, MAP kinase, and calcium. These characteristics suggest that PACAP may influence neuronal development. Here we have studied the effects of PACAP on mesencephalic dopaminergic neurons using primary cultures from embryonic rats. PACAP increased the number of tyrosine hydroxylase (TH)-immunoreactive neurons, elevated TH protein, and enhanced tritiated dopamine uptake in these cultures. Moreover, PACAP counteracted the effects of 6-hydroxydopamine treatments, which induce cell death of dopaminergic neurons. In situ hybridisation showed that both PACAP and PACAP receptor type 1 are present in developing and adult rat mesencephalon. These results show that PACAP has a neurotrophic action on dopaminergic neurons and partially protects them against 6-OHDA induced neurotoxicity. J. Neurosci. Res. 54:698–706, 1998. © 1998 Wiley-Liss, Inc.
Article
The pattern of retrograde axonal transport of the target-derived neurotrophic molecule, nerve growth factor (NGF), correlates with its trophic actions in adult neurons. We have determined that the NGF-related neurotrophins, brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), are also retrogradely transported by distinct populations of peripheral and central nervous system neurons in the adult. All three 125I-labeled neurotrophins are retrogradely transported to sites previously shown to contain neurotrophin-responsive neurons as assessed in vitro, such as dorsal root ganglion and basal forebrain neurons. The patterns of transport also indicate the existence of neuronal populations that selectively transport NT-3 and/or BDNF, but not NGF, such as spinal cord motor neurons, neurons in the entorhinal cortex, thalamus, and neurons within the hippocampus itself. Our observations suggest that neurotrophins are transported by overlapping as well as distinct populations of neurons when injected into a given target field. Retrograde transport may thus be predictive of neuronal types selectively responsive to either BDNF or NT-3 in the adult, as first demonstrated for NGF.
Article
To investigate the role of the gp140trk receptor tyrosine kinase in nerve growth factor (NGF)-induced differentiation, we have overexpressed gp140trk in the NGF-responsive PC12 cell line. Here we demonstrate that overexpression of gp140trk results in marked changes in NGF-induced differentiation. Whereas PC12 cells elaborated neurites after 2 days of continuous exposure to NGF, PC12 cells overexpressing gp140trk by 20-fold(trk-PC12) began this process within hours. Compared with wild-type PC12 cells, trk-PC12 exhibited an increase in both high and low affinity NGF-binding sites. Furthermore, trk-PC12 cells displayed an enhanced level of NGF-dependent gp140trk autophosphorylation, and this activity was sustained for many hours following ligand binding. The tyrosine phosphorylation or activity of several cellular proteins, such as PLC-gamma 1, PI-3 kinase, and Erk1 and the expression of the mRNA for the late response gene transin were also sustained as a consequence of gp140trk overexpression. The data indicate that overexpression of gp140trk in PC12 cells markedly accelerates NGF-induced differentiation pathways, possibly through the elevation of gp140trk tyrosine kinase activity.