Download Ciliary neurotrophic factor may activate mature

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Secreted frizzled-related protein 1 wikipedia , lookup

Neurotransmitter wikipedia , lookup

Molecular neuroscience wikipedia , lookup

NMDA receptor wikipedia , lookup

Index of biochemistry articles wikipedia , lookup

Biochemical cascade wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

Signal transduction wikipedia , lookup

Endocannabinoid system wikipedia , lookup

Clinical neurochemistry wikipedia , lookup

Paracrine signalling wikipedia , lookup

Transcript
Molecular and Cellular Neuroscience 17, 373–384 (2001)
doi:10.1006/mcne.2000.0926, available online at http://www.idealibrary.com on
MCN
Ciliary Neurotrophic Factor May Activate Mature
Astrocytes via Binding with the Leukemia
Inhibitory Factor Receptor
Christelle Monville,* Muriel Coulpier, † Luciano Conti, ‡
Claudio De-Fraja, ‡ Patrick Dreyfus,* Christiane Fages,*
Danielle Riche,* Marcienne Tardy,* Elena Cattaneo, ‡
and Marc Peschanski* ,1
*INSERM U421, IM3, Faculté de Médecine, 94010 Créteil cedex, France; †Regeneron
Pharmaceuticals, Inc., Tarrytown, New York 10591; and ‡Institute of Pharmacological
Sciences, 20133 Milan, Italy
Ciliary neurotrophic factor (CNTF) acts on immature astrocytes that express its trimeric receptor. In contrast,
mature astrocytes do not significantly express the specific CNTF␣ receptor subunit, yet they respond to CNTF
administration in vivo. Here we show that this controversy
may be solved by a shift in astroglial sensitivity to CNTF
over time, related to a change in the type of receptor
bound by the cytokine on mature astrocytes. A convergent set of results supports the hypothesis that the CNTF
effect is due to the illegitimate binding on the leukemia
inhibitory factor receptor (LIFR): (i) it requires high concentration of recombinant rat CNTF; (ii) it involves the
Jak/Stat and Ras-MAPK pathways; (iii) it is preserved in
CNTFR␣ⴚ/ⴚ cells; (iv) it is potentiated by soluble CNTFR␣
added to the medium; and (v) it is significantly decreased
by a partial antagonist of LIFR. On these bases, we propose a mechanistic model in which, in the adult brain, a
CNTF/LIFR interglial system may be modulated by neurons that synthesize CNTFR␣.
INTRODUCTION
Synthesis of the ciliary neurotrophic factor (CNTF)
and of all three components of its trimeric receptor is
widespread in the adult central nervous system
(Squinto et al., 1990; Stöckli et al., 1991; Sendtner et al.,
1994; MacLennan et al., 1996). Its neuroprotective ef1
To whom correspondence and reprint requests should be addressed at INSERM U421, IM3, Faculté de médecine, 8 rue du Général
Sarrail, 94010 Créteil cedex, France. Fax: 33 1 49 81 37 09. E-mail:
[email protected].
1044-7431/01 $35.00
Copyright © 2001 by Academic Press
All rights of reproduction in any form reserved.
fects, together with the source of the factor in astrocytes,
the location of its specific CNTFR␣ receptor subunit
specifically on neurons in vivo, and its lack of a signal
peptide, have led to the hypothesis that CNTF may act
as an “injury molecule” (Thoenen, 1991; Sendtner et al.,
1997), protecting neurons after release from astrocytes
following injury. Recent demonstration of phenotypic
changes in astrocytes after administration of CNTF into
the adult brain (Winter et al., 1995; Levison et al., 1996;
Clatterbuck et al., 1996; Lisovoski et al., 1997) has suggested that it may also play a role in inducing astroglial
activation.
The CNTF triggers astroglial differentiation very efficiently in glial precursors and immature astrocytes
(Hughes et al., 1988; Lillien et al., 1988; Kahn et al., 1995;
Bonni et al., 1997). This effect is likely related to the
binding of the cytokine to its receptor, the three components of which are expressed by these precursors and
immature cells (Bonni et al., 1997). In contrast, effects of
CNTF on mature astrocytes have remained controversial. Astroglial activation in long-term cultures treated
with CNTF has indeed been altogether weak or nonexisting (Meyer and Unsicker, 1994; Smith et al., 1996). In
addition, despite the observation of CNTF effects in
vivo, astrocytes in the adult brain have not been shown
to express the specific ␣ subunit of the trimeric CNTF
receptor (Squinto et al., 1990; MacLennan et al., 1996;
Kordower et al., 1997), the association of which to the
dimeric receptor of the leukemia inhibitory factor (LIF)
is necessary to form the trimeric CNTF receptor (Ip et
al., 1993; Davis et al., 1993b). Paradoxically, this expres-
373
374
sion is observed by Western and Northern blotting in
long-term astrocytic cultures in which the effects of
CNTF are not solid (Rudge et al., 1994; Alderson et al.,
1999).
These data have led to different hypotheses, including the expression of an undetectable, though sufficient,
amount of CNTFR␣ in adult astrocytes (Levison et al.,
1996) or the need for an interaction with another cell
type to obtain the CNTF-elicited astroglial activation
(Kahn et al., 1997). The present study has explored an
alternative hypothesis that these apparently discrepant
data may reveal a shift in astroglial sensitivity to CNTF
over time, related to a change in the type of receptor
bound by the cytokine. This hypothesis was based upon
studies that have demonstrated that CNTF may act on
cells lacking the CNTFR␣ subunit via illegitimate binding with the dimeric LIF receptor, although at a much
higher concentration (Schooltink et al., 1992; Gearing et
al., 1994). The present results show that CNTF-induced
astroglial activation can indeed be observed in longterm astroglial cultures and that the biological characteristics of this effect are compatible with the illegitimate binding of CNTF with the dimeric LIF receptor.
RESULTS
Enriched astroglial cell cultures were readily obtained from cerebral hemispheres of Swiss mice (Iffa
Credo, France), CNTF⫺/⫺ mice (BRL, Basel Switzerland), or CNTFR␣⫺/⫺ (Regeneron, Tarrytown, NY),
and maintained for several weeks without signs of cell
alteration.
CNTF Effect on Long-Term Astrocytes: Dose
Dependence and Intracellular Signaling Pathways
In immature astrocytes (maintained 7 days in culture,
not at confluence), addition of recombinant rat CNTF
(rCNTF) to the medium at concentrations as low as 10
ng/ml (lowest concentration used) provoked an increase in the cellular content of glial fibrillary acid
protein (GFAP) (Fig. 1a). Western blotting showed no
endogenous CNTF in these short-term cultures either in
the absence of treatment or 3 days after addition of
rCNTF to the medium.
In sharp contrast, rCNTF was inefficient at concentrations as high as 100 ng/ml in long-term cultures
when astrocytes were maintained in culture before
treatment until confluence was reached (around 14
days). An increase in the cellular content of GFAP was
observed in a significant and reproducible fashion only
Monville et al.
when the highest concentration of rCNTF studied (250
ng/ml) was used (Fig. 1b). Similarly, cellular content of
endogenous CNTF, which was quantifiable in these
more mature astrocytes, was increased significantly
above control levels only when 250 ng/ml of rCNTF
was added to the medium (Fig. 1c) and not following
treatment with lower doses of the cytokine. To eliminate an artifactual synergistic effect of the serum component TGF␤, a pan-TGF␤ antibody was added to the
culture medium. This treatment had no effect on the
increase of CNTF intracellular content after addition of
250 ng/ml of rCNTF to the medium.
A quantification of rCNTF, which would have been
internalized by cultured cells, was excluded by measuring no CNTF in samples obtained from CNTF⫺/⫺
mice after treatment of mature astrocytes with 250
ng/ml of rCNTF. CNTF cellular content was then used
as a marker of astroglial activation in following experiments, rather than GFAP, because this molecule is
present in astroglial cultures only after 14 days, not at 7,
and therefore may be a discriminant marker of mature
astrocytes. Consequently, using CNTF as a marker allowed us to overcome the potential problem created by
the persistence of a small subpopulation of astroglial
precursors or immature cells—which express GFAP but
not CNTF—in long-term cultures (see below). The two
intracellular signaling systems studied, namely the Jak/
STAT and the Ras-MAPkinase pathways, exhibited
rapid and sustained activation when long-term astroglial cultures were treated with 250 ng/ml of rCNTF.
Stat3 (Figs. 2a and 2b) and Stat1 (Figs. 2c and 2d) were
indeed strongly phosphorylated as soon as 5 min after
treatment and up to 1 h. In parallel, the adapter protein
Shc, ERK1, and ERK2, which are part of the Ras-MAPK
pathway associated with the CNTF receptor, were activated as soon as 5 min after rCNTF application (Figs.
3a and 3b). For these elements of the Ras-MAPK pathway, the activation was still significant 240 min after
application. In contrast, application of 30 ng/ml of rCNTF either was inefficient or induced only limited
phosphorylation.
Dependence of the Effects of CNTF on the
Presence or Concentration of CNTFR␣
The requirement for the presence of CNTFR␣ in the
astroglial effects of rCNTF at high concentration was assessed by adding the cytokine to the medium of cultures
of mature astrocytes obtained from CNTFR␣ knock-out
mice. In these cultures, addition of 250 ng/ml of rCNTF
induced an increase in CNTF cellular content similar to
that seen in wild-type cells (Fig. 4a), demonstrating that
CNTF May Act on Astrocytes via the LIF Receptor
375
the specific ␣ receptor subunit was not required in the
effects. The effect of providing CNTFR␣ to wild-type
mature astrocytes was then assessed using the soluble compound myc-sCNTFR␣. Under these conditions, a potentiation of the effects was observed as a
major shift toward values lower than the dose required for rCNTF effects on mature astrocytes. Increase of CNTF cellular content was indeed observed
with the lowest concentration studied (10 ng/ml) and
maintained at higher concentrations (Fig. 4b).
All three CNTF receptor subunits, including
CNTFR␣, were present in the long-term astroglial cultures (Fig. 6b). Immunocytochemical staining, however,
demonstrated that CNTFR␣ was not evenly distributed
in long-term cultures of astrocytes but was instead
present only on a small subpopulation of small bipolar
cells. The vast majority of cells which were large and
displayed the typical flattened morphology of longterm cultured astrocytes were not decorated by antibodies against CNTFR␣ (Fig. 5). In contrast, these latter
cells, which were the only ones to contain CNTF, expressed the two other subunits of the CNTF receptor,
LIFR␤ and gp130 (Fig. 5).
To check that an indirect mechanism was not involved
in the CNTF effects on mature astrocytes in which
CNTFR␣ present on the small cells would be donated to
the mature astrocytes, the concentration of CNTFR␣ was
modified in two different experiments. A mechanical lesion was made in the cultures to provoke an increase in
the concentration of CNTFR␣ (Fig. 6b). The lesion provoked an increase in CNTF cellular content at baseline but
did not amplify the effects of rCNTF (Fig. 6a). Reciprocally, a phosphatidylinositol-specific phospholipase C
(PIPLC) was used to cleave the glycosylphosphatidylinositol anchor that links CNTFR␣ to the cell membrane,
and the medium was changed before the application of
rCNTF to eliminate as much of the ␣ receptor subunit as
possible. The PIPLC treatment, the biological activity of
FIG. 1. (a) Effect of rCNTF on GFAP cellular content in immature
astrocytes. rCNTF provoked an increase of 44 ⫾ 9% (factorial
ANOVA significant at 95%, t test ***P ⬍ 0.001, **P ⬍ 0.01, or *P ⬍
0.05) at the lowest concentration used (10 ng/ml), compared to
untreated cultures. This effect was maintained, without further in-
crease, at all other concentrations. (b) Effect of rCNTF on GFAP
cellular content in mature astrocytes. Addition of 250 ng/ml rCNTF
induced a statistically significant increase compared to untreated
controls (⫹23 ⫾ 5.6%, factorial ANOVA significant at 95%, t test *P ⬍
0.05). In contrast, application of rCNTF provoked a statistically significant decrease, compared to untreated controls, for 30 and 50
ng/ml (⫺24 ⫾ 8.5 and ⫺16 ⫾ 1.7%, respectively, factorial ANOVA
significant at 95%, t test *P ⬍ 0.05). (c) Effect of rCNTF on CNTF
cellular content in mature astrocytes. No change in CNTF cellular
content was observed when rCNTF was added at 10, 30, 50, and 100
ng/ml concentrations. In contrast, a significant increase was consistently measured at 250 ng/ml (⫹39.6 ⫾ 10%, factorial ANOVA significant at 95%, t test **P ⬍ 0.01).
376
Monville et al.
FIG. 2. (a, b) Activation of the Jak/Stat pathway by rCNTF in mature astrocytes. Stat3 was phosphorylated with 30 ng/ml (⫹700 ⫾ 250%
of control, factorial ANOVA significant at 95%, t test *P ⬍ 0.05), and more strongly with 250 ng/ml (⫹3400 ⫾ 1800% of control, factorial
ANOVA significant at 95%, t test *P ⬍ 0.05) after 5 and 30 min (⫹297 ⫾ 86 and ⫹615 ⫾ 170% of control, factorial ANOVA significant at
95%, t test P ⬍ 0.01 in both cases). This activation was not observed after 60 min. (c, d) Stat1 was phosphorylated with 30 ng/ml (⫹140 ⫾
90%, NS) and 250 ng/ml (⫹409 ⫾ 160% of control, factorial ANOVA significant at 95%, t test *P ⬍ 0.05) after 5 and 30 min (⫹192 ⫾ 100%
with 30 ng/ml and ⫹321 ⫾ 170% with 250 ng/ml).
377
CNTF May Act on Astrocytes via the LIF Receptor
a molecule that was shown to partially block the activation of VIP by CNTF (Vernallis et al., 1997), was
added to the culture medium together with 250 ng/ml
of rCNTF. Under these conditions, a significant decrease (of about 50%) in the effects of rCNTF on astrocytes was observed (Fig. 7).
DISCUSSION
The main result of this study is that CNTF can provoke biochemical changes in mature astrocytes in vitro,
as previously observed in immature glial cells. The
receptor system is not, however, the complete tripartite
CNTF receptor since it does not involve the specific ␣
subunit. A convergent body of facts suggests that CNTF
may act on mature astrocytes by binding illegitimately
to the dimeric receptor of the LIF. This demonstrates a
developmental shift in astrocytes, since CNTF binds to
its own full trimeric receptor system on immature astrocytes and glial precursors. In the mature brain, CNTF
is synthesized by astrocytes and its specific CNTFR␣
subunit is synthesized—and potentially released in the
extracellular space via cleavage of its GPI anchor— by
neurons. Taking into account the present data, we propose a mechanistic model in which neurons may consequently play a role in the activation of mature astrocytes by modulating the efficacy of a CNTF/LIFR
interglial signaling system.
CNTF May Effect Biochemical Differentiation of
Mature Astrocytes via Binding to the LIF Receptor
FIG. 3. (a, b) Activation of the Shc-Grb2-MAPKs pathway by rCNTF
in mature astrocytes. ERK1 (p42MAPK) and mainly ERK2
(p44MAPK) were strongly activated with 250 ng/ml after 5 and 30
min (⫹470 ⫾ 60 and ⫹2133 ⫾ 490% of control, factorial ANOVA
significant at 95%, t test P ⬍ 0.001 in both cases).
which was controlled by observing an increase in cholinesterase activity in the medium, did not alter the effects
of rCNTF on mature astrocytes (Fig. 6a).
Dependence of the Effects of CNTF on the Activity
of the Dimeric LIF Receptor
To test the hypothesis that CNTF effects on astrocytes
require the activity of the dimeric LIF receptor, hLIF05,
CNTF provokes astroglial differentiation of glial precursors and strongly activates the synthesis of GFAP in
immature astrocytes, in vivo as well as in vitro (Hughes
et al., 1988; Lillien et al., 1988; Kahn et al., 1995, 1997;
Bonni et al., 1997; Rajan and McKay, 1998). In contrast,
an effect of CNTF on the synthesis of GFAP or endogenous CNTF has been observed in the adult only when
the cytokine was administered into the brain of living
rodents (Winter et al., 1995; Levison et al., 1996; Clatterbuck et al., 1996; Lisovoski et al., 1997) and not in purified astroglial cultures (Meyer and Unsicker, 1994;
Smith et al., 1996). These paradoxical results had led to
the hypothesis that activation of mature astrocytes by
CNTF was indirect and followed an effect of the cytokine on neighboring cells in vivo (Meyer and Unsicker,
1994; Smith et al., 1996). The present results do not
support this hypothesis since an activation of GFAP
and endogenous CNTF synthesis was obtained in longterm purified astroglial cultures. In a previous report, in
378
FIG. 4. (a) Effect of rCNTF on astrocytes obtained from CNTFR␣⫺/⫺
mice. Addition of 250 ng/ml induced an increase of CNTF cellular
content in both wild-type and CNTFR␣-deficient mice (⫹85 ⫾ 21 and
⫹143 ⫾ 47%, respectively, factorial ANOVA significant at 95%, t test
䡠䡠P ⬍ 0.01 and *P ⬍ 0.05). (b) Addition of soluble CNTFR␣ provoked a
major shift in the concentration of rCNTF required to observe glial
effects. Addition of sCNTFR␣ at 200 ng/ml to the culture medium was
followed by a major increase of CNTF cellular content with 10 ng/ml
(⫹66.5 ⫾ 30%, factorial ANOVA significant at 95%, t test *P ⬍ 0.05). A
similar increase was maintained, whatever the concentration of rCNTF
used, between 30 and 250 ng/ml.
which astroglial activation was observed in the brain of
adult rats following intracerebral injection of adenoviral vectors recombinant for a secretable form of CNTF
Monville et al.
(Lisovoski et al., 1997), the very restricted areas in which
this activation was observed around sites of potential
delivery of the cytokine had led us to hypothesize either
that CNTF did not spread freely far away from these
sites or that a high concentration of the cytokine was
required. The present in vitro results strongly support
the latter hypothesis since, in sharp contrast to the
results obtained in short-term cultures (of immature
astrocytes), there was no effect of CNTF observed at
concentrations lower than 10 ⫺8 M. This dose requirement explains the failure of previous in vitro studies to
observe the effects (Meyer and Unsicker, 1994; Smith et
al., 1996).
The existence of an effect of CNTF on mature astrocytes is paradoxical because all studies of the sites
of synthesis of the specific ␣ subunit of the CNTF
receptor, through visualization of mRNA (Ip et al.,
1993) or protein (MacLennan, 1996; Kordower et al.,
1997), have failed to reveal an astrocytic localization
in the adult brain. CNTFR␣ is, essentially, if not
exclusively, synthesized by neurons in the central
nervous system. To explain the effects of CNTF on
astrocytes, authors have hypothesized a low, undetectable level of CNTFR␣ expression (Levison et al.,
1996) or the activation of its expression in response to
inflammatory mechanisms (Rudge et al., 1994, 1995).
The present results support an alternative hypothesis
that CNTF may bind illegitimately to the dimeric LIF
receptor. CNTF normally binds to a high-affinity receptor complex, which is composed of three subunits,
CNTFR␣ and the two ␤ subunits (LIFR␤ and gp130),
which together form the receptor of another cytokine,
the leukemia inhibitory factor (Squinto et al., 1990;
Davis et al., 1993b; Ip et al., 1993; Stahl et al., 1994). It
has been shown, however, that CNTF can provoke
biological effects on cells that express only the LIF
receptor (Schooltink et al., 1992; Davis et al., 1993b;
Gearing et al., 1993). The arguments in favor of the
existence of such a mechanism in the present experiments with mature astrocytes are numerous: the astrocytic activation was observed at the same concentration of the cytokine when cells were taken from
mice that do not express CNTFR␣ or from wild-type
mice, indicating that this specific subunit is not required; although CNTFR␣ is present in long-term
astroglial cultures (Rudge et al., 1994; Alderson et al.,
1999), immunohistochemical analysis indicated that
it was not expressed by mature astrocytes, and alteration of its concentration did not modify the conditions of the astroglial effects of CNTF; the binding of
CNTF to the glial receptor provoked the phosphorylation of molecules that belong to the two intracellu-
CNTF May Act on Astrocytes via the LIF Receptor
379
FIG. 5. Immunocytochemical staining showing that the CNTFR␣ subunit is only present in a subpopulation of smaller bipolar cells. The two
␤ components of the LIF receptor (LIFR␤ and gp130) as well as CNTF are, in contrast, present in large cells with a flat polygonal shape and
several processes. Scale bar: 10 ␮m.
lar signaling pathways linked to the LIF (and by
extension the CNTF) receptor, namely the Jak/Stat
and the (Shc-)Ras-MAPK pathways (see references
and discussion in Stahl and Yancopoulos, 1994; Stahl
et al., 1994; Boulton et al., 1994; Bonni et al., 1997;
Rajan and McKay, 1998); these effects were characterized by the need for a much lower concentration of
the cytokine when CNTFR␣ was added in a soluble
form to the medium (at least 100-fold), a result comparable to those previously described for cells treated
with CNTF that expressed only the LIF receptor
(Davis et al., 1993b; Panayotatos et al., 1994); and
partial blockade of the binding of CNTF to the LIFR
using a specific antagonist (hLIF05) significantly reduced the effects of CNTF on astrocytes.
Altogether, the shift in response to CNTF observed
during the maturation of astrocytes may be explained
by the loss of expression of the CNTFR␣ subunit, and
therefore of the high-affinity receptor present at early
stages, and the consequent use of the two ␤ subunits of
the LIF receptor that form a low-affinity receptor for
CNTF.
380
FIG. 6. (a) Modulation of CNTFR␣ availability in astroglial cultures.
A lesion, which increased the level of CNTFR␣, did not amplify the
effects of rCNTF on CNTF cellular content (⫹72 ⫾ 10.8% of control,
factorial ANOVA significant at 95%, t test **P ⬍ 0.01). PIPLC treatment in order to remove CNTFR␣ anchored at the cell membranes of
the small bipolar cells did not alter the effects of rCNTF on the mature
astrocytes (⫹97 ⫾ 21.4% vs ⫹84 ⫾ 47% of control, ANOVA significant
at 95%, t test 䡠䡠䡠P ⬍ 0.001). (b) Western blot demonstrating the
presence of the three components of the trimeric CNTF receptor
(LIFR␤, 200K mol wt; gp130, 130K mol wt; CNTFR␣, 80K mol wt) in
mature astroglial cultures from wild-type mice. NL, nonlesioned; L,
lesioned.
An Interglial Signaling System Which Is Potentially
under Neuronal Control in the Adult Brain
The biological significance of such a low-affinity receptor system for CNTF on mature astrocytes is but a
matter of speculation. It is intriguing, however, that
Monville et al.
astrocytes are the only known source of CNTF in the
adult brain (Stöckli et al., 1991; Sendtner et al., 1994).
Release of large amounts of CNTF in the extracellular
space will therefore affect astrocytes via a paracrine/
autocrine system in the same time it affects neighboring
neurons that express CNTFR␣ (Ip et al., 1993; MacLennan, 1996; Kordower et al., 1997). Following injury to
the brain parenchyma, release of CNTF by dying astrocytes will thus provoke activation of surviving astrocytes in the close vicinity of the lesion, as well as exert
a neuroprotective role as hypothesized by several authors (Thoenen, 1991; Sendtner et al., 1997).
The strong potentiation of the CNTF effects on astrocytes by soluble CNTFR␣ suggests the possibility of an
additional type of signaling system. CNTFR␣ is synthesized by neurons but it is linked to their membrane by
a GPI anchor (Davis et al., 1991), which can be cleaved
even in the intact brain since it is detected in a soluble
form in the cerebrospinal fluid (Davis et al., 1993a). The
presence of CNTFR␣ in a soluble form in the brain will
transform the low-affinity astroglial receptor system
into a high-affinity one and, as a consequence, will
allow much smaller amounts of CNTF to provoke biochemical changes in astrocytes. Our data therefore are
compatible with the hypothesis of a three-partner system in which astroglial release of CNTF would affect
neighboring astrocytes in a manner which would
strongly depend upon neuronal release of CNTFR␣
(Fig. 8). A CNTF-based interglial signal could thus be
modulated by neurons in the absence of a lesion provoking the release of massive amounts of the cytokine.
The physiological conditions under which such a system may operate are, however, not known and their
very existence may even be questioned. On the one
hand, and although release of GPI-anchored CNTFR␣
has been demonstrated for skeletal muscle following
denervation, physiological conditions under which
neurons may modulate a similar release are not known.
On the other hand, the issue of a potential physiological
release of CNTF by astrocytes (i.e., not following injury)
remains unsolved. CNTF lacks a signal peptide, indicating that it is not released via conventional exocytotic
mechanisms (Thoenen, 1991; Sendtner et al., 1994). It
could, however, be released via a nonconventional energy-dependent pathway (Kamiguchi et al., 1995) reminiscent of those demonstrated for the 18-kDa isoform
of bFGF and IL-1␤ (Rubartelli et al., 1990; Florkiewicz et
al., 1995). This has not been observed, however, in astroglial cultures, the conditioned medium of which
does not support the survival of ciliary ganglion cells
(Rudge et al., 1992) and does not provoke phosphorylation of appropriate intracellular signaling proteins
CNTF May Act on Astrocytes via the LIF Receptor
381
FIG. 7. Effect of partial blockade of the LIFR on the effects of rCNTF on mature astrocytes. Addition of the partial LIFR antagonist hLIF05 to
the culture medium provoked a 50% reduction of the increase of the intracellular CNTF levels provoked by the addition of 250 ng/ml of rCNTF
in mature astrocytes (ANOVA significant at 95%, t test **P ⬍ 0.01).
(Rudge et al., 1995). The answer to that question, of a
potential physiological release of CNTF by astrocytes in
the mature brain, will determine whether the interglial
signaling system defined in the present study is just
another injury mechanism or whether it operates, under
the control of neurons releasing CNTFR␣, under currently unknown physiological conditions.
EXPERIMENTAL METHODS
Astroglial cultures were prepared from cerebral
hemispheres of neonatal Swiss mice (Iffa Credo,
France), CNTF knockout mice (BRL, Switzerland), and
neonatal CNTFR␣ knockout mice (Regeneron Pharmaceutical Inc., U.S.A.). Cultures were either grown at
confluence for 14 days, thus defining “mature” cultures,
or maintained only for 7 days. In the latter case, identified as “immature” cultures, cells occupied less than
50% of the dish.
Culture Conditions
Astroglial cultures were prepared as previously described (Bardakdjian et al., 1979). After 7 or 14 days in
vitro, the medium (minimal essential medium containing 2 mM glutamine, essential amino acids, 0.03% glucose, penicillin–streptomycin) was removed and replaced with the same medium but without fetal calf
serum (FCS). Recombinant rat CNTF (Boehringer
Mannheim, Germany) (rCNTF) was then added to the
medium at a concentration of 0, 10 (4.4 ⫻ 10 ⫺10 M), 30
(1.3 ⫻ 10 ⫺9 M), 50 (2.2 ⫻ 10 ⫺9 M), 100 (4.4 ⫻ 10 ⫺9 M), or
250 ng/ml (10 ⫺8 M). The cells were collected 3 days
later. In preliminary studies, it was confirmed (Levison
et al., 1996) using tritiated thymidine that CNTF does
not provoke astroglial proliferation in in vitro cultures.
Some of the mature astroglial cultures, prepared as
described above, were mechanically lesioned using the
fine tip of a plastic pipette 18 days after astroglial plating. After lesion, the medium was removed and cultures were treated with 250 ng/ml of rCNTF or left
untreated. In other experiments, PIPLC (Boehringer)
was added at the concentration of 1 U/ml and the
medium was exchanged 24 h later. In half of the dishes,
rCNTF was then applied as described above. PIPLC
activity was checked by quantifying the acetylcholine
esterase activity in the supernatant, according to the
technique described by Ellman et al. (1964). In the experiments with soluble CNTFR␣, 200 ng/ml of mycsCNTFR␣ (kindly provided by Ralph Laufer, IRBM,
Italy) was added to the medium without FCS after 14
days in vitro. Thirty minutes later, rCNTF was added at
0, 10, 30, 50, 100, and 250 ng/ml. The cells were collected 3 days later.
To block a potential binding of CNTF on the dimeric
LIFR, hLIF05 (kindly provided by Dr. Ann Vernallis,
UK) was used. This compound is a specific antagonist
of the LIF receptor that has been shown in other cell
models to partially block the activation of the VIP gene
382
Monville et al.
FIG. 8. CNTF/LIF receptor as an interglial signaling system under neuronal control. This scheme presents a hypothesis in which the
CNTF-based glial signaling system is open to a neuronal partner which can strongly amplify the binding efficiency of the ligand by providing
the CNTFR␣ subunit and thus restore a high-affinity receptor system.
by CNTF (50% inhibition with 5 ␮g/ml of hLIF05; Vernallis et al., 1997). hLIF05 was added 12 h before rCNTF
(250 ng/ml), and the cells were collected 24 h after
rCNTF treatment.
In all cases, the medium was removed at the end of
the experiments and each dish was rinsed three times
with HBSS (Hanks’ balanced salt solution, Seromed,
Germany). The cells were collected by scraping into 62.5
mM Tris–HCl (pH 6.8), 2 mM EDTA, 2 mM phenylmethylsulfonyl fluoride, 0.5% Triton X-100, and 2.3%
sodium dodecyl sulfate.
Biochemical Analysis
Total protein content was determined by the method
of Lowry et al. (1951) with bovine serum albumin as a
standard. The proteins were analyzed by Western blotting under the conditions described by Laemmli (1970).
Briefly, samples were boiled for 5 min after addition of
10% glycerol, 5% mercaptoethanol (or 5% dithiothreitol
for the MAPK-P), and 5% bromophenol blue, and then
lysates were electrophoresed on 7.5% (LIFR␤, gp130,
CNTFR␣, Stat3-P, and Stat1-P), 10% (GFAP and MAPKP), or 12% (CNTF) SDS–polyacrylamide gels. Gels were
blotted on nitrocellulose, blocked for 1 h in 5% nonfat
dry milk in TBS-T (20 mM Tris, pH 7.5/500 mM NaCl/
0.1% Tween 20), and then probed overnight at 4°C with
the following antibodies: polyclonal anti-CNTF (R&D
Systems, UK, 1/500), polyclonal anti-CNTFR␣ (kindly
provided by Dr. A. J. MacLennan, University of Florida,
U.S.A., 1/1000), monoclonal anti-gp130 (Santa Cruz,
U.S.A., 1/1000), polyclonal anti-LIFR␤ (Santa Cruz,
1/500), monoclonal anti-p44/p42 MAP kinase (or antiphospho ERK1/ERK2, New England Biolabs, UK,
1/1000), polyclonal anti-ERK1 (Santa Cruz, 1/4000),
polyclonal anti-phospho-specific Stat3 (Tyr705) (New
England Biolabs, 1/1000), polyclonal anti-phospho-specific Stat1 (Tyr701) (New England Biolabs, 1/1000),
monoclonal anti-Stat3 (Santa Cruz, 1/1000), and polyclonal anti-Stat1 (Santa Cruz, 1/1000). After washing
with TBS-T, membranes were incubated with a horseradish peroxidase-conjugated donkey anti-rabbit sec-
CNTF May Act on Astrocytes via the LIF Receptor
ondary antibody (for CNTFR␣, LIFR␤, ERK1, phosphospecific Stat3, phospho-specific stat1, and stat1), a
mouse anti-goat antibody (for CNTF), or a sheep antimouse antibody (for gp130, p44/p42 MAP kinase, and
Stat3), followed by the enhanced chemiluminescent reaction (Amersham, Sweden), according to the manufacturer’s instructions. For GFAP, the sheets were incubated with a polyclonal anti-GFAP antibody
(Dakopatts, Denmark, 1/100) and revealed with antirabbit IgG coupled to 125I, according to a previously
described technique (Andres-Barquin et al., 1994). The
sheets were placed in contact with a RPN6 film (Amersham). The levels of CNTF and GFAP were measured
by densitometry and brought back to the total protein
laid down.
To limit variations in their processing, extracts from
the control and all experimental conditions were treated
in parallel on a single sheet for each specific experiment.
In addition, to evaluate the variability between specific
experiments, control extracts were subsequently reloaded on a single sheet and processed together. Statistical analysis used one-factor ANOVA and unpaired t
test.
Immunohistochemical Analysis
The medium was removed and cells were rinsed two
times with PBS. They were then fixed with cold methanol (⫺20°C) and probed 1 h at room temperature with
the following antibodies: polyclonal anti-CNTF (1/100),
polyclonal anti-CNTFR␣ (1/50), monoclonal anti-gp130
(1/100), and polyclonal anti-LIFR␤ (1/100). After washing with PBS, cells were exposed to secondary antibodies linked to TRITC.
ACKNOWLEDGMENTS
These studies were supported by INSERM and Association Française contre les Myopathies. The authors gratefully acknowledge help
from Ralph Laufer (IRBM P. Angeletti, Rome), John McLennan (University of Florida College of Medicine, Gainesville, FL), Austin Smith
(Center for Genome Research, Edimburg, UK), Ann Vernallis (School
of Health and Life Sciences, Aston, UK), Tom De Chiara and George
Yancopoulos (Regeneron, Tarrytown, NY), Sharon L. Juliano
(USUHS, Bethesda, MD), Philippe Hantraye (CNRS, Orsay, Italy),
and Brigitte Onténiente (INSERM, Créteil, Italy).
REFERENCES
Alderson, R. F., Pearsall, D., Lindsay, R. M., and Wong, V. (1999).
Characterization of receptors for ciliary neurotrophic factor on rat
hippocampal astrocytes. Brain Res. 818: 236 –251.
383
Andres-Barquin, P. J., Fages, C., Le Prince, G., Rolland, B., and Tardy,
M. (1994). Thyroid hormones influence the astroglial plasticity:
Changes in the expression of glial fibrillary acidic protein (GFAP)
and of its encoding message. Neurochem. Res. 19: 65– 69.
Bardakdjian, J., Tardy, M., Pimoule, C., and Gonnard, P. (1979).
GABA metabolism in cultured glial cells. Neurochem. Res. 4: 517–
527.
Bonni, A., Sun, Y., Nadal-Vicens, M., Bhatt, A., Frank, D. A., Rozovski,
I., Stahl, N., Yancopoulos, G. D., and Greenberg, M. E. (1997).
Regulation of gliogenesis in the central nervous system by the
JAK-STAT signalling pathway. Science 278: 477– 483.
Boulton, T. G., Stahl, N., and Yancopoulos, G. D. (1994). Ciliary
neurotrophic factor/leukemia inhibitory factor/interleukin 6/oncostatin M family of cytokines induces tyrosine phosphorylation of
a common set of proteins overlapping those induced by other
cytokines and growth factors. J. Biol. Chem. 269: 11648 –11655.
Clatterbuck, R. E., Price, D. L., and Koliatsos, V. E. (1996). Ciliary
neurotrophic factor stimulates the expression of glial fibrillary
acidic protein by brain astrocytes in vivo. J. Comp. Neurol. 369:
543–551.
Davis, S., Aldrich, T. H., Ip, N. Y., Stahl, N., Scherer, S., Farruggella,
T., DiStefano, P. S., Curtis, R., Panayotatos, N., Gascan, H., Chevalier, S., and Yancopoulos, G. D. (1993a). Released form of CNTF
receptor alpha component as a soluble mediator of CNTF responses. Science 259: 1736 –1739.
Davis, S., Aldrich, T. H., Stahl, N., Pan, L., Taga, T., Kishimoto, T., Ip,
N. Y., and Yancopoulos, G. D. (1993b). LIFR beta and gp130 as
heterodimerizing signal transducers of the tripartite CNTF receptor. Science 260: 1805–1808.
Davis, S., Aldrich, T. H., Valenzuela, D. M., Wong, V. V., Furth, M. E.,
Squinto, S. P., and Yancopoulos, G. D. (1991). The receptor for
ciliary neurotrophic factor. Science 253: 59 – 63.
Ellman, G. L., Courtney, K. D., Andres, V., and Featherstone, R. M.
(1964). A new and rapid colorimetric determination of acetylcholinesterase. Biochem. Pharmacol. 7: 88 –95.
Florkiewicz, R. Z., Majack, R. A., Buechler, R. D., and Florkiewicz, E.
(1995). Quantitative export of FGF-2 occurs through an alternative,
energy-dependent, non-ER/Golgi pathway. J. Cell. Physiol. 162:
388 –399.
Gearing, D. P., Ziegler, S. F., Comeau, M. R., Friend, D., Thoma, B.,
Cosman, D., Park, L., and Mosley, B. (1994). Proliferative responses
and binding properties of hematopoietic cells transfected with lowaffinity receptors for leukemia inhibitory factor, oncostatin M, and
ciliary neurotrophic factor. Proc. Natl. Acad. Sci. USA 91: 1119 –1123.
Hughes, S. M., Lillien, L. E., Raff, M. C., Rohrer, H., and Sendtner, M.
(1988). Ciliary neurotrophic factor induces type-2 astrocyte differentiation in culture. Nature 335: 70 –73.
Ip, N. Y., McClain, J., Barrezueta, N. X., Aldrich, T. H., Pan, L., Li, Y.,
Wiegand, S. J., Friedman, B., Davis, S., and Yancopoulos, G. D.
(1993). The alpha component of the CNTF receptor is required for
signaling and defines potential CNTF targets in the adult and
during development. Neuron 10: 89 –102.
Junier, M. P., Coulpier, M., Le Forestier, N., Cadusseau, J., Suzuki, F.,
Peschanski, M., and Dreyfus, P. A. (1994). Transforming growth
factor alpha (TGFalpha) expression in degenerating motoneurons
of the murine mutant wobbler: A neuronal signal for astrogliosis?
J. Neurosci. 14: 4206 – 4216.
Kahn, M. A., Ellison, J. A., Chang, R. P., Speight, G. J., and de Vellis,
J. (1997). CNTF induces GFAP in a S-100 alpha brain cell population: The pattern of CNTF-alpha R suggests an indirect mode of
action. Dev. Brain Res. 98: 221–233.
Kahn, M. A., Ellison, J. A., Speight, G. J., and de Vellis, J. (1995). CNTF
384
regulation of astrogliosis and the activation of microglia in the
developing rat central nervous system. Brain Res. 685: 55– 67.
Kamiguchi, H., Yoshida, K., Sagoh, M., Sasaki, H., Inaba, M., Wakamoto, H., Otani, M., and Toya, S. (1995). Release of ciliary neurotrophic factor from cultured astrocytes and its modulation by cytokines. Neurochem. Res. 20: 1187–1193.
Kordower, J. H., Yaping-Chu, and MacLennan, A. J. (1997). Ciliary
neurotrophic factor receptor alpha-immunoreactivity in the monkey central nervous system. J. Comp. Neurol. 377: 365–380.
Laemmli, U. K. (1970). Cleavage of structural proteins during the
assembly of the head of bacteriophage T4. Nature 227: 680 – 685.
Le Prince, G., Fages, C., Rolland, B., Nunez, J., and Tardy, M. (1991).
DBcAMP effect on the expression of GFAP and of its encoding
mRNA in astroglial primary cultures. Glia 4: 322–326.
Levison, S. W., Ducceschi, M. H., Young, G. M., and Wood, T. L.
(1996). Acute exposure to CNTF in vivo induces multiple components of reactive gliosis. Exp. Neurol. 141: 256 –268.
Lillien, L. E., Sendtner, M., Rohrer, H., Hughes, S. M., and Raff, M. C.
(1988). Type-2 astrocytes development in rat brain cultures is initiated by a CNTF-like protein produced by type-1 astrocytes. Neuron 1: 485– 494.
Lisovoski, F., Akli, S., Peltekian, E., Vigne, E., Haase, G., Perricaudet,
M., Dreyfus, P. A., Kahn, A., and Peschanski, M. (1997). Phenotypic
alteration of astrocytes induced by ciliary neurotrophic factor in the
intact adult brain, as revealed by adenovirus-mediated gene transfer. J. Neurosci. 17: 7228 –7236.
Lowry, O. H., Rosebrough, N. J., Farr, A. L., and Randall, R. J. (1951).
Protein measurement with the Folin phenol reagent. J. Biol. Chem.
193: 265–275.
MacLennan, A. J., Vinson, E. M., Marks, L., McLaurin, D. L., Pfeifer,
M., and Lee, N. (1996). Immunohistochemical localization of ciliary
neurotrophic factor receptor alpha expression in the rat nervous
system. J. Neurosci. 16: 621– 630.
Meyer, V., and Unsicker, K. (1994). Cell density and exogenous CNTF
affect CNTF mRNA levels in glial cell cultures. Neuroreport 5:
1946 –1948.
Panayotatos, N., Everdeen, D., Liten, A., Somogyi, R., and Acheson,
A. (1994). Recombinant human CNTF receptor alpha: Production,
binding stoichiometry, and characterization of its activity as a
diffusible factor. Biochemistry 33: 5813–5818.
Raivich, G., Bohatschek, M., Kloss, C. U., Werner, A., Jones, L. L., and
Kreutzberg, G. W. (1999). Neuroglial activation repertoire in the
injured brain: Graded response, molecular mechanisms and cues to
physiological function. Brain Res. Brain Res. Rev. 30: 77–105.
Rajan, P., and McKay, R. D. (1998). Multiple routes to astrocytic
differentiation in the CNS. Neuroscience 18: 3620 –3629.
Rubartelli, A., Cozzolino, F., Talio, M., and Sitia, R. (1990). A novel
secretory pathway for interleukin-1␤, a protein lacking a signal
sequence. EMBO J. 9: 1503–1510.
Rudge, J., Li, Y., Pasnikowski, E., Mattson, K., Pan, L., Yancopoulos,
Monville et al.
G. D., Wiegand, S. J., Lindsay, R. M., and Ip, N. Y. (1994). Neurotrophic factor receptors and their signal transduction capabilities in
rat astrocytes. Eur. J. Neurosci. 6: 693–705.
Rudge, J. S., Alderson, R. F., and Pasnikowski, E. (1992). Expression of
ciliary neurotrophic factor and the neurotrophins—Nerve growth
factor, brain derived neurotrophic factor and neurotrophin-3 in
cultured rat hippocampal astrocytes. Eur. J. Neurosci. 4: 459 – 471.
Rudge, J. S., Pasnikowski, E. M., Holst, P., and Lindsay, R. M. (1995).
Changes in neurotrophic factor expression and receptor activation
following exposure of hippocampal neuron/astrocyte cocultures to
kainic acid. J. Neurosci. 15: 6856 – 6867.
Schooltink, H., Stoyan, T., Roeb, E., Heinrich, P. C., and Rose-John, S.
(1992). Ciliary neurotrophic factor induces acute-phase protein expression in hepatocytes. FEBS Lett. 314: 280 –284.
Sendtner, M., Carroll, P., Holtmann, B., Hughes, R. A., and Thoenen,
H. (1994). Ciliary neurotrophic factor. J. Neurobiol. 25: 1436 –1453.
Sendtner, M., Gotz, R., Holtmann, B., and Thoenen, H. (1997). Endogenous ciliary neurotrophic factor is a lesion factor for axotomized
motoneurons in adult mice. J. Neurosci. 17: 6999 –7006.
Smith, G. M., Hale, J., Pasnikowski, E. M., Lindsay, R. M., Wong, V.,
and Rudge, J. S. (1996). Astrocytes infected with replication-defective adenovirus containing a secreted form of CNTF or NT3 show
enhanced support of neuronal populations in vitro. Exp. Neurol. 139:
156 –166.
Squinto, S. P., Aldrich, T. H., Lindsay, R. M., Morrissey, D. M.,
Panayotatos, N., Bianco, S. M., Furth, M. E., and Yancopoulos, G. D.
(1990). Identification of functional receptors for ciliary neurotrophic
factor on neuronal cell lines and primary neurons. Neuron 5: 757–
766.
Stahl, N., Boulton, T. G., Farruggella, T., Ip, N. Y., and Davis, S. (1994).
Association and activation of Jak-Tyk kinases by CNTF-LIF-OSMIL-6 beta receptor components. Science 263: 92–95.
Stahl, N., and Yancopoulos, G. D. (1994). The tripartite CNTF receptor
complex: Activation and signaling involves components shared
with other cytokines. J. Neurobiol. 25: 1454 –1466.
Stöckli, K. A., Lillien, L. E., Nahernoe, M., Breitfeld, G., Hughes, R. A.,
Raff, M. C., Thoenen, H., and Sendtner, M. (1991). Regional distribution, developmental changes, and cellular localization of CNTFmessenger RNA and protein in the rat brain. J. Cell Biol. 115:
447– 459.
Thoenen, H. (1991). The changing scene of neurotrophic factors.
Trends Neurosci. 14: 165–170.
Vernallis, A. B., Hudson K., and Heath J. K. (1997). An antagonist for
the leukemia inhibitory factor receptor inhibits leukemia inhibitory
factor, cardiotrophin-1, ciliary neurotrophic factor, and oncostatin
M. J. Biol. Chem. 272: 26947–26952.
Winter, C. G., Saotome, Y., Levison, S. W., and Hirsh, D. (1995). A role
for ciliary neurotrophic factor as an inducer of reactive gliosis, the
glial response to central nervous system injury. Proc. Natl. Acad. Sci.
USA 92: 5865–5869.
Received February
Revised October
Accepted October
Published online January
29,
16,
25,
19,
2000
2000
2000
2001