Vous êtes sur la page 1sur 10

The Journal of Immunology

Semaphorin CD100 from Activated T Lymphocytes Induces


Process Extension Collapse in Oligodendrocytes and Death of
Immature Neural Cells1

Pascale Giraudon,2,3* Peggy Vincent,2* Carine Vuaillat,2* Olivier Verlaeten,2* Luis Cartier,‡
Anne Marie-Cardine,† Mireille Mutin,* Armand Bensussan,† Marie-Françoise Belin,* and
Laurence Boumsell†
An inappropriate cross talk between activated T lymphocytes infiltrating the CNS and neural cells can sustain the onset and
progression of demyelination and axonal degeneration in neuroinflammatory diseases. To mimic this deleterious cross talk, we
designed an experimental paradigm consisting of transient cocultures of T lymphocytes chronically activated by retrovirus in-
fection (not virus productive) with human multipotent neural precursors or primary oligodendrocytes from rat brain. We showed
that activated T lymphocytes induced apoptotic death of multipotent neural progenitors and immature oligodendrocytes after a
progressive collapse of their process extensions. These effects were reminiscent of those induced by brain semaphorin on neural
cells. Blockade by specific Abs of soluble CD100 (sCD100)/semaphorin 4D released by activated T cells, or treatment with
rsCD100, demonstrated that this immune semaphorin has the ability to collapse oligodendrocyte process extensions and to trigger
neural cell apoptosis, most likely through receptors of the plexin family. The specific presence of sCD100 in the cerebrospinal fluid
and of CD100-expressing T lymphocytes in the spinal cord of patients suffering with neuroinflammatory demyelination pointed to
the potential pathological effect of sCD100 in the CNS. Thus, our results show that CD100 is a new important element in the
deleterious T cell-neural cell cross talk during neuroinflammation and suggest its role in demyelination or absence of remyelination
in neuroinflammatory diseases including multiple sclerosis and human T lymphotropic virus type 1-associated myelopathy. The
Journal of Immunology, 2004, 172: 1246 –1255.

I nteraction between the immune and the nervous systems may more complete understanding of the functional properties of di-
trigger an inappropriate cross talk, as suggested by demyeli- verse molecules either in normal or pathological conditions. It is
nation and axonal loss in neuroinflammatory diseases. In at- becoming clear that semaphorins, which are well known for their
tempting to understand this cross talk, two key issues need to be role in axonal steering, synapse formation, and chemorepulsion in
resolved: the entry and survival of activated T lymphocytes in the the developing nervous system (2– 4), also have roles in the im-
brain, and the identification of immune molecules leading to de- mune system. In fact, CD100 (also known as semaphorin 4D
myelination. A large and diverse molecular repertoire shared by (SEMA-4D))4 and SEMA-4A, two immune semaphorins, are ex-
immune and CNS, such as cytokines, chemokines, the metallopro- pressed constitutively in T cells (5, 6), function as soluble ligand
teinase/inhibitor, and Fas/Fas ligand systems, revealing remark- to regulate the humoral and cellular immune responses (7–9), and
able similarities between signaling molecules and cellular recep- are suspected to be involved in autoimmune diseases such as ex-
tors in both systems, could participate in this cross talk (1). It also perimental autoimmune encephalitis (EAE) (6, 10). SEMA-3A
provides essential information reflecting important steps toward a acts in axonal guidance, apoptosis of neural precursors or neurons,
and oligodendrocyte alteration (11–14), and inhibits the chemo-
kine-induced migration of human immune cells similarly to
*Institut National de la Santé et de la Recherche Médicale Research Unit 433, Ex- CD100 (15). Neuropilin-1, a component of SEMA-3A receptor in
perimental Neurobiology and Physiopathology, Federative Institut of Neuroscience brain, is also expressed in immune cells and plays a role in the
19 Faculty of Medicine R Laennec, Lyon, France; †Institut National de la Santé et de
la Recherche Médicale Research Unit 448, Differentiation, Interaction, Activation, establishment of contacts between naive T lymphocytes and den-
and Migration of Human Immune Cells, Faculty of Medicine, Créteil, France; and dritic cells (16). In this context, we propose that soluble CD100

Departement of Neuroscience, Faculty of Medicine, Hospidal del Salvador, San- (sCD100), which is produced upon T lymphocyte activation
tiago, Chile
through proteolysis by a metalloproteinase (17), could be respon-
Received for publication May 19, 2003. Accepted for publication October 30, 2003.
sible for an inappropriate response in neural-immune interactions
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
during inflammation.
with 18 U.S.C. Section 1734 solely to indicate this fact. Neuroinflammatory diseases, including multiple sclerosis (MS)
1
This work was supported by grants from French Agencies of Research on Multiple and myelopathy associated with human T lymphotropic virus type
Sclerosis and on HIV (Agence Recherche Selérose Plaques, Ligue Recherche Sclé- 1 (HTLV-1) infection (TSP/HAM), are characterized by inflam-
rose Plaques, Association Francaise Contre les Myopathies, and Agence Nationale de
Recherches sur le SIDA) and Institut National de la Santé et de la Recherche Médicale
mation associated with damage of the white matter and axonal
funds.
2
P.G., P.V., C.V., and O.V. contributed equally to this study. 4
Abbreviations used in this paper: SEMA, semaphorin; CNPase, cyclic nucleotide
3
Address correspondence and reprint requests to Dr. Pascale Giraudon, Unit 433 phosphodiesterase; CSF, cerebrospinal fluid; EAE, experimental autoimmune enceph-
Institut National de la Santé et de la Recherche Médicale, Faculty of Medicine R alitis; GalCer, galactocerebroside; GFAP, glial acidic fibrillary protein; HTLV-1, hu-
Laennec, rue G. Paradin 69372 Lyon CEDEX 08 France. E-mail address: giraudon@ man T lymphotropic virus type 1; MS, multiple sclerosis; sCD100, soluble CD100;
lyon.inserm.fr TSP/HAM, tropical spastic paraparesis/HTLV-1-associated myelopathy.

Copyright © 2004 by The American Association of Immunologists, Inc. 0022-1767/04/$02.00


The Journal of Immunology 1247

degeneration in the brain and spinal cord (18 –20). Both infiltrated Alternatively, in the same set of experiments, the neural cells were
T lymphocytes and inflammatory mediators are suspected to par- treated with rsCD100 from the supernatant of permanently transfected Ju-
ticipate in pathogenic mechanisms (21–25). The persistent demy- rkat cells, used crude or after purification on anti-CD100 BB18-mAb col-
umn (1 ng/␮l concentration). Supernatant from Jurkat cells transfected to
elination in these diseases and the reported sensibility of neural produce CD27 was used in similar condition as control. Each experiment
progenitors to inflammatory mediators suggest the dysfunction or was repeated at least three times.
death of the precursors or myelinating oligodendrocytes required
for remyelination (26 –28). Axonal loss, detected early in the dis- Patients
ease by magnetic resonance spectroscopy (29, 30), could result The CNS tissue samples from three TSP/HAM patients and two nonin-
from defects in remyelination (31) and abnormal levels of molec- fected patients (Parkinson disease, car crash) were examined for CD100-
ular signals that regulate axon extension such as semaphorins (32). expressing T cells. Paraffin-embedded spinal cords were examined using
H&E-safran and Luxol Fast Blue for myelin detection. Clinical and patho-
Assuming that immune semaphorins produced by infiltrating T
logical characteristics of these TSP/HAM patients are demonstrated in Ta-
lymphocytes may have a deleterious effect on neural cells, we ble I. The CSF from patients suffering with TSP/HAM (9 patients), men-
investigated the in vitro effect of activated T cells releasing ingitis (4 patients), or encephalomyelitis (3 patients) were examined for the
sCD100-SEMA-4D and rsCD100 protein on human pluripotent presence of sCD100 by a sandwich ELISA, as previously described (5). All
neural precursors and on rat oligodendrocytes. These cellular con- TSP/HAM patients tested were positive for HTLV-1 provirus.
tacts mimic interactions occurring during neuroinflammation be-
Immunodetection
tween infiltrating/activated T lymphocytes and oligodendrocytes
or pluripotent neural precursors still capable of generating glial Oligodendrocytes were identified in the rat primary glial culture by immu-
nofluorescence on culture slide or by flow cytometry (5, 35), with anti-
cells in adult brain (33). In respect to the effects of brain sema-
CNPase (Sigma-Aldrich, St. Louis, MO), anti-GalCer, anti-NG2 (Chemi-
phorins on neural precursors and oligodendrocytes (12, 13), we con International, Temecula, CA), anti-myelin-associated glycoprotein
particularly focused on process extensions and cell survival. Next, (Boehringer Mannheim, Indianapolis, IN), and anti-myelin basic protein
the in vivo functional relevance was examined by looking for: 1) (Serotec, Oxford, U.K.) mAbs. Astrocytes were identified in the same con-
the presence of sCD100 in the cerebrospinal fluid (CSF), and 2) the ditions with a rabbit polyclonal Ab (anti-GFAP; DAKO, Carpenteria, CA).
Human fetal neural precursors were detected with anti-␤3-tubulin (T8660;
presence of cell surface CD100 on activated T lymphocytes in Sigma-Aldrich). Semaphorin receptors were detected with polyclonal anti-
postmortem spinal cords from patients suffering with neuroinflam- human plexin-B1 (N18; SantaCruz-Tebu, Le Perray, France) and anti-
matory demyelination (TSP/HAM). MAM neuropilin-1-blocking polyclonal Ab (12). Immune cells were de-
tected in spinal cord sections from TSP/HAM patients by
immunofluorescence with anti-CD4 (M0716; DAKO), anti-CD8 (M7103;
Materials and Methods DAKO), anti-CD45RO (UCHL1; DAKO), and anti-fascin (M3567;
Model of T lymphocyte-neural cell interaction DAKO) mAbs. Membrane-bound CD100 was detected with an anti-CD100
polyclonal antiserum directed against the intracellular portion of the pro-
The following CD4⫹ T cells were used: 1) the human CD25⫹CD100⫹ T
tein (aa 799 – 813). After dewaxing with toluene and ethanol, sections were
cell line C8166, which is activated by chronic retroviral infection (HTLV-I,
incubated in blocking solution (1% BSA, 0.3% Triton X-100, 1 h), then
no virus productive) (34) and released high level of sCD100 (3 ng/␮l from
with specific Abs (4°C/overnight). Alexa546-labeled anti-mouse or Al-
20 ⫻ 106 cells in 1 ml compared with 1 ng/␮l from CD100-transfected
exa488-labeled anti-rabbit Ig Abs (Molecular Probes, Eugene, OR) were
Jurkat T cells in same conditions), as detected by ELISA (5); 2) the non-
then applied.
activated CD4⫹ T cell line CEM used as control; and 3) the primary culture
of T lymphocytes isolated from a patient infected with HTLV-1 (CIB,
CD25⫹, CD69⫹, CD100⫹). These T lymphocytes were transiently cocul- RNA detection
tured, as previously described (35), with the following human and rat neu- mRNA were detected by extraction, followed by RT-PCR and Southern
ral cells (ratio 1/10, respectively) growing on culture slide or in flask: 1) the blotting using appropriate [33P]dATP 5⬘ end-labeled internal oligonucleo-
human pluripotent neural precursor cell line Dev, which has the ability to tide probes, as previously described (38). Oligonucleotide primers were
differentiate into neurons, astrocytes, and oligodendrocytes (36, 37); 2) chosen from their mRNA sequences (GenBank access numbers HSU60800
primary culture of rat glial cells (38), which contained 35–54% oligoden- for CD100, NM002663 for plexin-B1, NM012401 for plexin-B2,
drocytes, the remaining cells corresponding to glial acidic fibrillary protein AF149019 for plexin-B3, L26081 for SEMA-3A, NM01101 for ␤-actin,
(GFAP)-positive astrocytes. The oligodendrocytes have distinct pheno- used as control). One set of nucleotide primers (forward, TGGTGAAGC
typic stages identified with a panel of specific Abs by immunochemistry or CAAGTGATGAG; reverse, CTGCAGAAGTTCGTGGATGA) was se-
flow cytometry (see below): early oligodendrocyte precursor (pre-oligo- lected for amplification of a common 300-bp amplicon in the three
dendrocyte) expressing the NG2 chondroitin sulfate (10 –15%), immature plexin-B mRNA.
oligodendrocyte expressing galactocerebroside (GalCer, 17–25%) and cy-
clic nucleotide phosphodiesterase (CNPase), mature oligodendrocyte ex- Apoptosis analysis
pressing myelin-associated glycoprotein (3–15%) and myelin basic pro-
tein, and 3) human fetal neural cells cultivated from the subventricular zone In each experiment, apoptotic death was detected by three methods: 1) the
identified by immunodetection of ␤3-tubulin. Following transient coculture TUNEL method performed on culture slide (Promega, Madison, WI); 2)
(20 h) of C8166 or CEM T lymphocytes with human or rat neural cells, the immunodetection of the executer caspase, active caspase-3 (rabbit serum;
nonadherent T cells were eliminated by washing of neural cultures, and BD PharMingen, San Diego, CA); and 3) detection of apoptotic bodies
their elimination was verified by the lack of CD4 Ag in flow cytometry. For with DNA intercal 4⬘,6⬘-diamidino-2-phenylindole (1 ␮g/ml; Sigma-
some experiments, these cocultures were treated with anti-CD100-purified Aldrich) staining of nucleus. Codetection of TUNEL, active caspase-3, and
Abs, namely the blocking sCD100 BD16 mAb or the nonblocking oligodendrocyte or astrocyte markers was also performed to identify the
BB18 mAb. damaged cell population within the glial culture.

Table I. Clinical and pathological characteristics of TSP/HAM patients

CNS Neuropathology

Case Age/Sex Duration of Illness Cause of Death Inflammation Myelin/axonal loss Atrophy

1 62 /F 15 years Pulmonary embolism ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹


2 35 /M 4 years Mesenteric thrombosis ⫹⫹ ⫹⫹⫹ ⫹⫹
3 65 /F 8 years Pneumonia ⫹ ⫹ ⫹
1248 IMMUNE SEMA IN LYMPHOCYTE-NEURAL CELL INTERACTION

Statistical analysis enhanced. The number of TUNEL-positive cells increased from


The number of total cells (nucleus staining with 4⬘,6⬘-diamidino-2-phe- 17.2 ⫾ 2.4 per field in C8166-treated neural precursors vs 7 ⫾ 2.2
nylindole), TUNEL, and active caspase-3-positive cells was counted in in CEM-treated and 4.5 ⫾ 2.7 in untreated cells ( p ⬍ 0.001).
neural precusors and primary glial cells grown in culture slide and tran- Apoptotic bodies in TUNEL-positive cells are shown in Fig. 1A. In
siently cocultured with C8166 or CEM T cells (15–20 microscope fields/ addition, the executer caspase, active caspase-3, was detected by
800-1000 cells counted per experiment, 3 independent experiments) using
AnalySiS 3.2 software. The values were expressed as mean ⫾ SEM of immunofluorescence. As shown in Fig. 1B (three independent ex-
positive cells per field. Cell loss was calculated by counting the total cell periments gave similar results), active caspase-3 was detected in
number per field in each experimental situation or by flow cytometry after 25.5 ⫾ 8.8 cells per field following contact with C8166 T cells vs
immunodetection of oligodendrocyte populations. Differences between 11.3 ⫾ 3.7 in CEM-treated and 10.8 ⫾ 1.7 in untreated neural
groups were calculated with the Student’s t test. Measure of number and
length of oligodendrocyte process was performed in the cocultured glial precursors. Increase in cell apoptosis corroborated the decrease in
culture by using the same software (3 independent experiments). cell number (Fig. 1C). At 48 h postcontact with C8166 T cells,
there was 24 ⫾ 4.4% neural cell loss and 58.5 ⫾ 9.2% at 72 h, p ⬍
Results 0.001. Interestingly, neural precursor death was observed at a same
CD100-producing T cells dramatically affect neural precursor extent following treatment with culture supernatant of CD100-pro-
and oligodendrocyte integrity ducing T cells, C8166, or coculture without contact (Transwell
The effect of immune semaphorin SEMA-4D/CD100 released chamber) with these T cells. Thus, in contrast to T cell control,
from chronically activated T lymphocytes was investigated on hu- CD100-producing T cells dramatically enhanced the rate of natural
man pluripotent neural precursors (Dev cells) and on rat primary death in neural precursors, probably via a soluble factor.
oligodendrocytes within glial primoculture by analyzing neural The relevance to this observation was confirmed with additional
cell morphology and survival after contact. The T cell line C8166, cellular models: 1) primary culture of human fetal neural cells
which is activated by chronic retroviral infection and produces originating from the subventricular zone in contact with CD100-
high level of sCD100, was transiently cocultured with these neural producing C8166 cells; 2) neural precursor cell line Dev in contact
cells. The non-sCD100-producing T cells, CEM, were used as con- with primary T cells (CIB) activated by retroviral infection
trol. Spontaneous apoptosis was detected in 0.41–5.96% of un- (CD25⫹, CD69⫹, CD100⫹). Human fetal precursors did not sur-
treated Dev cells. Following contact with CD100-producing T vive following contact with C8166 T cells, while contact with
cells (48 h), their level of spontaneous apoptotic death was greatly CEM T cell did not change their survival levels (data not shown).

FIGURE 1. CD100-induced death in human neural precursors. A–C, Apoptotic death detected at 48 h by the TUNEL method (A), and estimated by
counting the number of active caspase-3-positive cells (B) and the total cell numbers (C) in human neural precursor cells transiently cocultured (20 h) with
control or CD100-producing T lymphocytes (mean ⫾ SEM cell number per field, microscopy AnalySiS, Student’s t test, representative experiment). D–F,
Similar analysis on neural precursors following 48-h treatment with rsCD100 protein (0.5 ng/␮l rsCD100 purified on CD100 BB18-mAb column).
The Journal of Immunology 1249

C8166 and primary T cells induced apoptosis in Dev cells at a (13.3 ⫾ 0.3 per field vs 6.3 ⫾ 0.3 in CEM-treated culture and
similar level (6.9, 10.3, and 7.1% of apoptotic cells detected at 27, 6 ⫾ 0.86 in untreated culture; Fig. 3A) and active caspase-3-
48, and 72 h, respectively, following contact with C8166 vs 2.9, positive cells (19.7 ⫾ 8.1 per field vs 7.3 ⫾ 5.5 in CEM-treated
3.5, and 5.2% after contact with CIB). culture; data not shown). As shown in Fig. 3B, apoptosis re-
The effect of CD100-producing T cells was also examined on rat sulted in a partial loss in oligodendrocyte population, evidenced
glial primary culture containing oligodendrocytes at various stages by the reduced number of oligodendrocytes per field (3.7 ⫾ 0.7
of maturation and astrocytes. Transient coculture with C8166 T oligodendrocytes per field vs 6.9 ⫾ 0.5 in CEM-treated glial
cells induced cell damages of oligodendrocytes, but not of astro- culture and 6.7 ⫾ 0.8 in untreated culture) and the total oligo-
cytes, which remained unchanged in number and morphology. In dendrocyte number (26.8 ⫾ 3.6% loss at 48 h following contact
fact, a progressive collapse and loss of process extensions were with C8166, p ⬍ 0.01). The number of astrocytes in the same
observed in glial cells identified as immature oligodendrocytes by treated cultures was not changed (10.7 ⫾ 0.4, 12.0 ⫾ 0.9, and
immunodetection of the specific marker, GalCer (Fig. 2). The 12.0 ⫾ 0.9 astrocytes per field, respectively) (Fig. 3B). In par-
length and number of process extensions of these immature oligo- allel experiments using astrocytes purified from rat glial culture
dendrocytes were progressively reduced after 24- and 48-h contact or established in cell line (C8S), CD100-producing T lympho-
with CD100-producing T cells CEM (Fig. 2B) compared with cul- cytes never induced astrocyte death (data not shown).
ture treated with control T cell (Fig. 2A) or untreated culture (data Interestingly, cytometry analysis performed on glial culture
not shown). The relative measure of the length (1 ⫾ 0.2 ␮m vs following contact with CD100-producing T cells showed a dra-
2.1 ⫾ 0.3 ␮m in CEM-treated glial culture at 48 h) and the count matic decrease in the number of immature oligodendrocytes
of processes (1.3 ⫾ 0.2 process per cell vs 5.3 ⫾ 0.6 in CEM- (Fig. 3C) identified by immunodetection of GalCer (38 ⫾ 2.4%
treated glial culture at 48 h) were estimated in three independent of total oligodendrocytes vs 58.6 ⫾ 6.6% in coculture with
experiments (Fig. 2D). Contact with CD100-producing T cells in- CEM), while population of early oligodendrocyte progenitors,
duced collapse of cell process in immature oligodendrocytes, identified by NG2-specific marker, was not significantly mod-
while the control T cells had no effect. Interestingly, the T cell- ified (47.8 ⫾ 14.1% of total oligodendrocytes vs 30.6 ⫾ 1.3 in
induced collapse of oligodendrocyte processes was followed by coculture with CEM). In addition, double labeling of GalCer
a late apoptotic death detected in the glial culture. As observed and TUNEL identified immature oligodendrocytes as the dying
in three independent experiments, contact with CD100-produc- cells (Fig. 3D). Collectively, these effects are reminiscent of the
ing T cells increased the number of TUNEL-positive cells previously reported collapse of oligodendrocyte processes and

FIGURE 2. CD100-induced alteration of oligoden-


drocyte process. Oligodendrocytes detected in the rat
primary glial culture by GalCer immunodetection (A).
Morphological changes in GalCer-positive cells at 24
and 48 h following transient coculture with CD100⫹ T
cells (B) or treatment with rsCD100 protein (C). No
change after coculture with control T cells (A). D,
Number and length of GalCer-positive cell process ex-
tensions after 24- and 48-h coculture with T cells or
treatment with rsCD100 measured using microscopy
AnalySiS (mean ⫾ SEM of process number and
length, 10 –20 measures, 3 experiments).
1250 IMMUNE SEMA IN LYMPHOCYTE-NEURAL CELL INTERACTION

FIGURE 3. CD100 induced the death of


immature oligodendrocytes. A, Count of ap-
optotic (TUNEL⫹) cells in cocultured or
rsCD100-treated glial culture (mean ⫾
SEM positive cell number per field). B,
Number of total oligodendrocytes and as-
trocytes in glial culture 48 h after coculture
with control and CD100-producing T cells
or treatment with rsCD100 (mean ⫾ SEM
cell number per field). C, Flow cytometry
analysis of oligodendrocyte populations
(percentage of total cell number) detected
in rat glial culture by immunodetection of
GalCer (immature oligodendrocytes) and
NG2 (early oligodendrocyte progenitors)
after coculture with CD100-producing T
cells. D, Double labeling of GalCer and
TUNEL identifying immature oligodendro-
cytes as dying cells.

death of neural precursors induced by secreted SEMA-3A, a apoptotic cells, as shown by active caspase-3 expression (15.0 ⫾
brain-derived semaphorin (12–14). 2.3 positive cells per field in culture treated with 0.5 ng/␮l
rsCD100 vs 6.0 ⫾ 1.9 positive cells in untreated culture; data not
sCD100 released from activated T lymphocytes causes the shown) and the number of TUNEL-positive cells (13.4 ⫾ 0.8 per
deleterious effect of T cell contact field vs 6.0 ⫾ 0.86 in untreated culture; Fig. 3A). Such an increase
We next looked at whether CD100 was involved in the T cell- in apoptosis marker expression corroborated the decreased number
induced neural cell damages. The fact that T cell supernatant re- of total oligodendrocytes under sCD100 treatment (3.6 ⫾ 0.6 oli-
produced the effect of T cell contact on neural cells led us to godendrocytes per field vs 6.7 ⫾ 0.8 in untreated culture; Fig. 3B)
suspect sCD100 as the deleterious factor. The direct effect of and resulted in 29 ⫾ 4% loss of oligodendrocytes at 48 h post-
CD100 was demonstrated by treating the neural precursor cells treatment. By contrast, there was no alteration of astrocyte survival
with rsCD100, released as a dimer from transfected Jurkat cells in the same cultures as shown in Fig. 3B. In addition, increased
(15). Similar to the contact with CD100-producing T cells (Fig. 1, doses of rsCD100 (0.1, 1, and 5 ng/␮l) induced apoptosis in a
D–F), treatment with rsCD100 induced apoptotic death of neural dose-dependent manner (7.5 ⫾ 0.7, 13.3 ⫾ 1.8, and 16.6 ⫾ 1.5
precursors. Increase in the number of active caspase-3-positive
cells was detected in rsCD100-treated neural precursors (25.3 ⫾
1.6 positive cells per field in culture treated with 0.5 ng/␮l
rsCD100 vs 10.8 ⫾ 1.8 in untreated culture; Fig. 1E) and was
associated with a decreased cell number per field (Fig. 1F). A
prominent cell loss (43.5 ⫾ 13.4%) was next observed at 72 h.
Treatment with increasing doses of rsCD100 (0.01, 0.1, and 0.5
ng/␮l) induced apoptosis of neural precursors in a dose-dependent
manner (7.8 ⫾ 1.2, 19.8 ⫾ 3, and 21.8 ⫾ 0.2 active caspase-3-
positive cells per field, respectively, vs 6.4 ⫾ 1.2 in culture treated
with control supernatant; Fig. 4A).
Treatment of glial primary culture with rsCD100 also resulted in
morphological evidence of cell damage in oligodendrocytes (Fig.
2). This treatment reduced the number of processes (1.4 ⫾ 0.2
FIGURE 4. Dose-dependent effect of CD100 on neural cells. Evaluation
processes per oligodendrocyte at 48 h vs 5.3 ⫾ 0.4 in untreated of apoptotic cell number expressing active caspase-3 in neural precursors
culture; Fig. 2D) and their relative length (1 ⫾ 0.2 ␮m at 48 h vs (A) or glial primary culture (B) following treatment by increasing doses of
2.1 ⫾ 0.3 ␮m in untreated culture) in immature oligodendrocytes rsCD100 (0.01, 0.1, and 0.5 ng/␮l) (mean ⫾ SEM active caspase-3 cells
identified by GalCer immunodetection (Fig. 2C). This morpholog- per field). rsCD100 was collected from CD100-transfected Jurkat cells and
ical deterioration was followed, at 48 h, by an increased number of control supernatant (sControl) from CD27-transfected Jurkat cells.
The Journal of Immunology 1251

active caspase-3-positive cells per field, respectively, vs 6.0 ⫾ 0.8 Receptors of the plexin family may be involved in the sCD100
in untreated glial culture; Fig. 4B). Double labeling of the glial effect on neural cells
culture with oligodendrocyte markers and TUNEL identified im-
Semaphorins may signal in neural cells through receptors of the
mature oligodendrocytes (GalCer positive) as a cell population
neuropilin and plexin families (39). Neuropilins act as coreceptors
sensitive to sCD100 treatment (data not shown). Thus, rsCD100
with A plexins, while B plexins alone behave as fully functional
induced a deleterious effect on neural precursors and oligodendro-
signal transducers for both transmembrane and secreted forms of
cytes similarly to CD100-producing T cells, while no detectable
change was observed on astrocytes. Higher rsCD100 dose (1 ng/ semaphorins. Our previous work had shown that neuropilin-1 is
␮l) induced the death of 64% neural precursors and 37% total present on human neural precursors and mediates SEMA-3A-in-
oligodendrocytes at 72 h. In control experiments, treatment of duced apoptosis of these cells (12). To eliminate the involvement
these human and rat neural cells with a supernatant from Jurkat of neuropilin-1 and SEMA-3A in the T cell-induced apoptosis of
stably transfected with an unrelated molecule, CD27, did not in- neural precursors, we added anti-MAM neuropilin-1 Ab in the T
duce any change (Fig. 4A), further demonstrating that this delete- neural cell coculture. This Ab, previously shown to block the
rious effect was attributable to sCD100. SEMA-3A-induced neural precursor death (12), had no significant
Finally, involvement of CD100 in the T cell-mediated damage effect on the rate of death induced by CD100-producing T cells
on neural cells was confirmed by treatment of cocultures with (Fig. 6A). In addition, the remote possibility of an effect of anti-
CD100-specific Abs. The anti-CD100 BD16 mAb, previously CD100 BD16 mAb on SEMA-3A activity (15) was ruled out by
shown to inhibit activity of sCD100 (15), was able to antagonize the absence of SEMA-3A expression in T cells, as shown by RT-
the C8166 T cell-induced effects on neural precursors and imma- PCR analysis of SEMA-3A mRNA in neural and T cells (Fig. 6B).
ture oligodendrocytes (Fig. 5). In fact, BD16 mAb used at increas- These observations excluded the implication of neuropilin-1/
ing concentrations progressively decreased, in a dose-dependent SEMA-3A in the T cell-mediated damages. Involvement of plex-
manner, the number of apoptotic neural precursors detected by the ins, in particular plexin-B1, identified as a receptor for sCD100 in
TUNEL method at 48 h postcontact with CD100-producing T cells human cells (39), was next investigated. The presence of mRNA
(13.4 ⫾ 1.4, 9.9 ⫾ 0.2, 2.3 ⫾ 0.6, and 3.2 ⫾ 0.8 positive cells per coding for plexin-B1, -B2, or -B3 in human neural precursors and
field, respectively, vs 16.5 ⫾ 0.9 in mAb-untreated coculture; Fig.
human fetal cortex (used as positive control) was evidenced by
5A). In addition, BD16 treatment reduced the T cell-mediated cell
RT-PCR. Taking into account the high homology of RNA se-
loss estimated at 72 h (41.5 ⫾ 0.7% cell loss vs 62.0 ⫾ 7.1% in
quence between these three plexins, oligomers were selected for
mAb-untreated coculture). In contrast, anti-CD100 BB18 mAb,
their ability to amplify a common 300-bp amplicon. Amplicons
which has no ability to block sCD100 activity, did not reduce T
cell-mediated damage (data not shown). Similarly, treatment of the were further identified by restriction enzymes, PstI and Sau3AI,
glial primary cells with BD16 mAb along with the coculture of which can generate 171- and 129-bp products in the plexin-B1 and
CD100-producing T cells reduced the T cell-mediated loss of oli- plexin-B3 amplicon, and 205- and 95-bp products in the plexin-B1
godendrocytes. As shown in Fig. 5B, BD16 treatment normalized and plexin-B2 amplicon, respectively. PstI and Sau3AI generated the
the number of oligodendrocytes per field. BD16 treatment also expected products from a 300-bp amplicon (Fig. 6C), showing the
decreased the number of TUNEL-positive cells compared with un- presence of plexin-B-coding mRNA in human neural precursors as in
treated coculture (Fig. 5, C3 vs C2). Thus, BD16 mAb exerted a human fetal cortex. The presence of plexin-B1 at the cell membrane
protective effect toward immature oligodendrocytes as exemplified was confirmed by immunodetection on live neural precursors, and
by the number of CNPase-positive cells higher in coculture under flow cytometry detected 50 –54% positive cells (Fig. 6D). The pos-
Ab treatment (Fig. 5, C3 vs C2). sible involvement of plexin-B1 in the CD100-producing T cell-me-

FIGURE 5. Reduction of the T cell-mediated dam-


age by CD100-blocking BD16 mAb. Neural precursors
(A) and glial culture (B and C) were cocultured with
control T cells, or with CD100⫹ T cells and treated or
not treated with anti-CD100 BD16 mAb (0.1, 0.4, 1, 2
ng/␮l for neural precursors, 1 ng/␮l for glial culture).
Apoptotic cells were detected by the TUNEL method,
and positive cell number was evaluated (mean ⫾ SEM
positive cells per field, microscopy AnalySiS). A, Re-
duction in a dose-dependent manner of T cell-mediated
apoptosis in neural precursors by BD16 mAb. B, Count
of astrocytes and oligodendrocytes in glial culture
(mean ⫾ SEM cell number per field, microscopy Analy-
SiS): decrease in oligodendrocyte number after cocul-
ture with CD100-producing T cells (B2) and resettled
number under BD16 mAb treatment (B3). No modifi-
cation in astrocyte population. C, A view of apoptosis
detection by the TUNEL method (green) and of imma-
ture oligodendrocyte population by CNPase immunode-
tection (red) in glial primary culture. Increase in the
number of TUNEL-positive cells after coculture with
CD100-producing T cells (C2) compared with coculture
with control T cells (C1) and reduction under BD16
mAb treatment (C3). In parallel, decrease in the number
of CNPase-positive cells (C2) and limited reduction un-
der BD16 mAb treatment (C3).
1252 IMMUNE SEMA IN LYMPHOCYTE-NEURAL CELL INTERACTION

FIGURE 6. Involvement of semaphorin receptor plexin-B in CD100-mediated effect on neural cells. A, Number of apoptotic/active caspase-3-positive
neural precursor cells in the presence of CD100-producing T cells with or without anti-plexin-B1 and anti-neuropilin-1 Abs (mean ⫾ SEM active
caspase-3-positive cells per field, microscopy AnalySiS). Reduction of T cell-mediated apoptosis by anti-plexin-B1 Ab. B, Detection by RT-PCR and
Southern blotting using [33P]dATP 5⬘ end-labeled internal probes of mRNA coding for SEMA-3A and ␤-actin (as housekeeping gene) in: 1) glial primary
culture; 2) neural precursors; 3) neural precursors ⫹ CD100⫹ T cells; and 4) CD100⫹ T cells. T cells did not express SEMA-3A. C, Detection by RT-PCR
of mRNA coding for a 300-bp amplicon common to plexin-B1, plexin-B2, and plexin-B3 in human neural precursors and human fetal brain. Identification
of the subsequent generation of 171- and 129-bp products by PstI in plexin-B1 and -B3 amplicon, and of 205- and 95-bp products by Sau3AI in plexin-B1
and -B2 amplicon. mRNA coding for plexin-B was present in neural precursors as in fetal brain. D, Plexin-B1 detected in neural precursor cells by
immunofluorescence and flow cytometry analysis (54% positive).

diated apoptosis in neural precursors was suggested by results of treat- CD45RO, or anti-fascin Abs identified the infiltrating immune
ment with anti-plexin-B1 Ab. When added throughout the coculture, cells as primarily CD45RO-positive cells and T lymphocytes with
anti-plexin-B1 Ab reduced the number of apoptotic neural cells (Fig. a predominance of CD8 T cells (Fig. 7B, case 1). As BD16 and
6A) and the rate of cell loss (5 vs 25.4% in untreated coculture at 48 h) BB18 anti-CD100 mAbs did not react with fixed cells, we devel-
induced by CD100-producing T cells. Treatment of rat oligodendro- oped a new polyclonal rabbit Ab recognizing an intracellular pep-
cytes cocultured with CD100-producing T cells did not modify the tide of the CD100 molecule to determine whether the membrane-
rate of cell apoptosis (data not shown), probably due to the human bound CD100 was detected on infiltrated immune cells. Double
specificity of this anti-plexin Ab. labeling using anti-CD100 and anti-CD45RO revealed the coex-
pression of these molecules in cell from the hemopoietic lineage.
sCD100- and CD100-expressing T cells in patients with The highest density of double-labeled cells was found in meningia
neuroinflammatory disorder (Fig. 7C) and around blood vessels (Fig. 7, E and F). Double-
The functional relevance of these experimental data was tested by labeled T lymphocytes were also observed in parenchyma (Fig.
examining the presence of sCD100 in CSF and CD100-positive 7F) both in gray and white matters. Astrocytosis in the white mat-
infiltrating T lymphocytes in postmortem CNS from patients suf- ter and around blood vessels was revealed by GFAP immunostain-
fering with demyelination associated with neuroinflammation, ing (Fig. 7G) of spinal cord from patient with high inflammation
TSP/HAM. CD100 was detected in spinal cord and CSF from level (case 1). Interestingly, we had detected the matrix metallo-
these patients, but not from patients with noninflammatory neuro- proteinase-9 within astrocytosis in the spinal cord of this patient
logical injuries (Table II and Fig. 7). Three patients suffering with (35). In contrast, the few CD45RO-positive cells present in spinal
TSP/HAM and exhibiting various levels of neuroinflammation ev- cords of nonneuroinflammatory patients were all CD100 negative
idenced by immune infiltrates were chosen for the histological (two cases studied; data not shown). Assuming that the expression
study. Anatomopathology analysis of paraffin-embedded spinal and release of sCD100 may be enhanced within the inflammatory
cords, using standard staining (H&E-safran), Luxol Fast Blue for neural tissue and CSF by high levels of metalloproteinases in TSP/
myelin detection, and immunofluorescence for cell identification, HAM patients (40, 41), the expression of sCD100 was analyzed in
indicated that spinal cords from these patients displayed demyeli- the CSF from patients with TSP/HAM (n ⫽ 9) or patients with
nation (Fig. 7A) associated with moderate to marked meningial nonneuroinflammatory diseases (n ⫽ 7) by ELISA. This analysis
(Fig. 7, A–C) and perivascular (Fig. 7, D and E) infiltrates of im- revealed the presence of sCD100 at a level of 97.7 ⫾ 23.1 ng/ml
mune cells. Level of neuroinflammation correlated with the extent in CSF from TSP/HAM patients (Table II), while levels were un-
of atrophy in the thoracic level and degeneration of the lateral detectable in the CSF from other groups of patients.
corticospinal and spinocerebellar tracts, the diffuse loss of myelin
and axons, and the thickness of the leptomeninges and astrocyto- Discussion
sis, as previously observed (19) (also see Table I). Immunofluo- The current pathogenesis sequence of inflammatory demyelinating
rescence analysis of spinal cords with anti-CD4, anti-CD8, anti- diseases, such as MS, starts with an inflammatory phase, which
The Journal of Immunology 1253

Table II. sCD100 detected by ELISA in the CSF from patients infected
with HTLV-1 and suffering with demyelination associated with virus-
induced neuroinflammation (TSP/HAM)a

TSP/HAM Patients CD100 (ng/ml)

1 132.6 ⫾ 29.9
2 66.6 ⫾ 14.2
3 158.3 ⫾ 17.2
4 79.6 ⫾ 36.3
5 123.9 ⫾ 42.7
6 60 ⫾ 9.9
7 62.9 ⫾ 14.2
8 57.9 ⫾ 16.3
9 75 ⫾ 27.6
a
Patients with other neurological diseases (n ⫽ 7) were negative for CD100
detection in CSF.

fulfills the criteria of an autoimmune disease, followed by a phase


of selective demyelination, and finally a neurodegenerative phase
(42– 44). The initial event of inflammation is the migration of ac-
tivated T lymphocytes of Th1 phenotype to the brain and spinal
cord, where they release deleterious proinflammatory cytokines.
There is indeed in vivo evidence that Th1-related cytokines such as
TNF-␣ and IFN-␥ are involved in lesion formation (22, 25, 45).
However, heterogeneity with respect to clinical course and re-
sponse to therapy has led to the concept of heterogeneous patho-
genic mechanisms of demyelination (46). Additional demyelinat-
ing amplification factors are probably required to produce the large
demyelinating lesions detected in patients. The present study
points out the deleterious effect of the immune semaphorin CD100
on oligodendrocyte and neural precursor integrity and proposes
that immune semaphorins are candidates in the pathogenic mech- FIGURE 7. Detection of CD100-expressing T cells in the spinal cord of
anism of demyelination. patients. Examination of spinal cords from three patients infected with
HTLV-1 and suffering with demyelination associated with virus-induced
We show that sCD100, reported to be highly expressed in ac-
neuroinflammation (TSP/HAM). A and D, H&E-safran staining and Luxol
tivated T lymphocytes and released from the T cell surface through
Fast Blue for myelin detection (histology) showed demyelination (A, open
a metalloproteinase-mediated proteolytic cleavage (17), induced a areas in blue staining, open arrow) and immune cell infiltration (filled
progressive decrease in process extensions of immature oligoden- arrow) in meningia (A) and around blood vessels (D). B, Immunodetection
drocytes, followed by their death, and the death of human multi- of the T cell markers CD4 and CD8 in meningia. E, C, and F, Immuno-
potent neural precursors. Thus, sCD100 of immune origin can ini- detection of CD100 (green) and CD45RO (red) in meningia (C), around
tiate a signaling cascade impairing neural precursor and blood vessel (E), and in parenchyma (F). Arrow triangle ⫽ coexpression
oligodendrocyte homeostasis, similarly to the neural SEMA-3A (orange); arrow square ⫽ monoexpression. Moderate to marked infiltration
(12, 13, 47). Interestingly, sCD100 affected mainly the immature is seen in F: F1, case 1; F2, case 2; F3, case 3. Astrocytosis identified by
oligodendrocytes (GalCer positive), while oligodendrocyte precur- GFAP immunodetection (G, case 1).
sors (NG2 positive) were preserved from T cell-mediated effect
and proliferate. This observation suggests the presence of a win-
dow of vulnerability to CD100 in immature oligodendrocytes and blocking mAb, BD16, to reduce the damage to neural cells
not in precursors. In fact, NG2-positive oligodendrocyte precur- mediated by activated T lymphocytes, in contrast to the nonblock-
sors are the principal dividing cell population within the perinatal ing BB18 mAb, confirms the physiological role of sCD100. In
and adult brain (48), but after a period of perinatal proliferation, addition, detection of sCD100 in the CSF of patients suffering with
GalCer-positive differentiating oligodendrocytes send a signal neuroinflammatory demyelination (TSP/HAM) and the presence
back to their precursors preventing their differentiation. The alter- of numerous infiltrating CD100/CD45RO-positive cells in their
ation of this feedback mechanism is thought to cause the rapid spinal cord, in contrast to patients suffering with noninflammatory
proliferation of NG2-positive cells during demyelinating disease. neurological injury, support the idea of a correlation between
In our model, the selective loss of GalCer-positive oligodendro- CD100 release and demyelination. The collapse of process exten-
cytes, the negative proliferation signal, following contact with sions in the immature/premyelinating oligodendrocyte population
sCD100-producing T lymphocytes, could explain the sustained and their death induced by sCD100 would dramatically compro-
presence of NG2-positive oligodendrocytes in the culture. The dif- mise the capacity of remyelination in inflamed brain. The present
ferential susceptibility of astrocytes and oligodendrocytes to data together with 1) the abundant expression of CD100 by acti-
CD100-induced damage is intriguing, as both cells express plexin- vated T cells (7, 10), 2) the elevated expression of metalloprotein-
B1. However, apoptosis in neural cells is strictly controlled by ases within the CNS of patients suffering neuroinflammatory de-
sequestration of active caspase, regulation of death receptor activ- myelination (23, 40), and 3) the gradual reduction in the size of the
ity, and expression of apoptosis-inhibitory proteins (49). Cell type- oligodendrocyte population, but the sustained presence of NG2-
specific regulations at these various levels could explain the dif- positive cells observed in brains of patients suffering neuroinflam-
ferential susceptibility to CD100. The capacity of sCD100- matory demyelination (27, 50), strongly suggest that infiltrated/
1254 IMMUNE SEMA IN LYMPHOCYTE-NEURAL CELL INTERACTION

activated T cells affect myelination through release of factors toxic axonal damage, and irreversible neurological disability is sus-
for neural cells, including semaphorin CD100. pected (19, 25, 42, 54).
In addition to its deleterious effects on neural cells, sCD100
could participate in the autoimmune response within the brain of Acknowledgment
patients suffering with neuroinflammatory demyelination (21, 51). We thank Isabelle Chabert de Ponnat for FACS analysis.
In fact, the two murine immune semaphorins, CD100 and SEMA-
4A, are expressed constitutively in T cells and function as soluble References
ligands to CD72 and Tim2, respectively, to regulate the humoral 1. Boulanger, L. M., G. S. Huh, and C. J. Shatz. 2001. Neuronal plasticity and
cellular immunity: shared molecular mechanisms. Curr. Opin. Neurobiol.
and cellular immune response (7–9). They are suspected to be 11:568.
involved in autoimmune diseases (6, 10, 52). sCD100 produced 2. Liu, B. P., and S. M. Strittmatter. 2001. Semaphorin-mediated axonal guidance
after lymphocyte activation has a potent costimulatory function to via Rho-related G proteins. Curr. Opin. Cell Biol. 13:619.
3. Kolodkin, A. L., D. J. Matthes, T. P. O’Connor, N. H. Patel, A. Admon,
induce proliferation of B cells and to enhance the maturation of D. Bentley, and C. S. Goodman. 1992. Fasciclin IV: sequence, expression, and
professional APCs (8, 53). This is reflected by the presence of function during growth cone guidance in the grasshopper embryo. Neuron 9:831.
4. Chisholm, A., and M. Tessier-Lavigne. 1999. Conservation and divergence of
significant levels of sCD100 in the serum of MLR/lpr mice devel- axon guidance mechanisms. Curr. Opin. Neurobiol. 9:603.
oping autoantibodies (10). In addition, mice carrying a truncated 5. Hall, K. T., L. Boumsell, J. L. Schultze, V. A. Boussiotis, D. M. Dorfman,
CD100 transgene encoding an easily released sCD100 develop A. A. Cardoso, A. Bensussan, L. M. Nadler, and G. J. Freeman. 1996. Human
CD100, a novel leukocyte semaphorin that promotes B-cell aggregation and dif-
EAE more rapidly than normal mice (52). The relation between ferentiation. Proc. Natl. Acad. Sci. USA 93:11780.
this autoimmune response to the CD100 transgene overexpression 6. Kumanogoh, A., S. Marukawa, K. Suzuki, N. Takegahara, C. Watanabe, E.
Ch’ng, I. Ishida, H. Fujimura, S. Sakoda, K. Yoshida, and H. Kikutani. 2002.
was proved by enhanced concentration of sCD100 in body fluids Class IV semaphorin Sema4A enhances T-cell activation and interacts with
and, conversely, by the fact that the CD100⫺/⫺ mice are resistant Tim-2. Nature 419:629.
to EAE. A direct role for sCD100 in oligodendrocyte pathogenesis 7. Bismuth, G., and L. Boumsell. 2002. Controlling the immune system through
semaphorins. Sci. STKE 2002:RE4.
remains to be investigated. More recently, the implication of 8. Suzuki, K., A. Kumanogoh, and H. Kikutani. 2003. CD100/Sema4D, a lympho-
SEMA-4A in the differentiation and activation of T cells upon cyte semaphorin involved in the regulation of humoral and cellular immune re-
sponses. Cytokine Growth Factor Rev. 14:17.
interaction with its receptor Tim2 was reported (6). Interestingly, 9. Kumanogoh, A., and H. Kikutani. 2001. The CD100-CD72 interaction: a novel
treating mice with anti-SEMA-4A mAb blocked the development mechanism of immune regulation. Trends Immunol. 22:670.
of EAE induced by the antigenic peptide derived from myelin 10. Wang, X., A. Kumanogoh, C. Watanabe, W. Shi, K. Yoshida, and H. Kikutani.
2001. Functional soluble CD100/Sema4D released from activated lymphocytes:
oligodendrocyte glycoprotein. Although the mAb effect was evi- possible role in normal and pathologic immune responses. Blood 97:3498.
dent when injected at an early phase of T cell responses against 11. Goshima, Y., T. Ito, Y. Sasaki, and F. Nakamura. 2002. Semaphorins as signals
for cell repulsion and invasion. J. Clin. Invest. 109:993.
myelin oligodendrocyte glycoprotein, it would be interesting to 12. Bagnard, D., C. Vaillant, S. T. Khuth, N. Dufay, M. Lohrum, A. W. Puschel,
look for a direct effect of SEMA-4A on oligodendrocytes. The fact M. F. Belin, J. Bolz, and N. Thomasset. 2001. Semaphorin 3A-vascular endo-
that SEMA-3A and sCD100 can inhibit the migration of immune thelial growth factor-165 balance mediates migration and apoptosis of neural
progenitor cells by the recruitment of shared receptor. J. Neurosci. 21:3332.
cells (15) also suggests that these semaphorins may affect migra- 13. Ricard, D., V. Rogemond, E. Charrier, M. Aguera, D. Bagnard, M. F. Belin,
tory T lymphocytes within the CNS. Taken together, these works N. Thomasset, and J. Honnorat. 2001. Isolation and expression pattern of human
Unc-33-like phosphoprotein 6/collapsin response mediator protein 5 (Ulip6/
and our findings in humans indicate that in neuroinflammatory CRMP5): coexistence with Ulip2/CRMP2 in Sema3a-sensitive oligodendrocytes.
situation, sCD100 and SEMA-3A could participate, through bind- J. Neurosci. 21:7203.
ing to immune and neural cells, in a close partnership, both in the 14. Shirvan, A., R. Shina, I. Ziv, E. Melamed, and A. Barzilai. 2000. Induction of
neuronal apoptosis by Semaphorin3A-derived peptide. Brain Res. Mol. Brain
sequestration and stimulation of immune cells within the CNS and Res. 83:81.
in the collapse of oligodendrocyte processes and neural precursor 15. Delaire, S., C. Billard, R. Tordjman, A. Chedotal, A. Elhabazi, A. Bensussan, and
L. Boumsell. 2001. Biological activity of soluble CD100. II. Soluble CD100,
death. similarly to H-SemaIII, inhibits immune cell migration. J. Immunol. 166:4348.
The presence of neuropilin-1 in neural precursors and oligoden- 16. Tordjman, R., Y. Lepelletier, V. Lemarchandel, M. Cambot, P. Gaulard,
drocytes and its involvement in the class 3 semaphorin-mediated O. Hermine, and P. H. Romeo. 2002. A neuronal receptor, neuropilin-1, is es-
sential for the initiation of the primary immune response. Nat. Immun. 3:477.
apoptosis of neural precursors and changes in oligodendrocytes 17. Elhabazi, A., S. Delaire, A. Bensussan, L. Boumsell, and G. Bismuth. 2001.
were previously reported (12, 13, 16). Our present data show that Biological activity of soluble CD100. I. The extracellular region of CD100 is
released from the surface of T lymphocytes by regulated proteolysis. J. Immunol.
neuropilin-1 is not the signaling receptor of sCD100 in neural 166:4341.
cells, as blocking anti-neuropilin-1 Ab failed to prevent CD100- 18. Prineas, J. W., R. O. Barnard, T. Revesz, E. E. Kwon, L. Sharer, and E. S. Cho.
mediated apoptosis. As B plexins fully function as signal trans- 1993. Multiple sclerosis: pathology of recurrent lesions. Brain 116:681.
19. Cartier, L. M., J. G. Cea, C. Vergara, F. Araya, and P. Born. 1997. Clinical and
ducers for both transmembrane and secreted forms of class 4 sema- neuropathological study of six patients with spastic paraparesis associated with
phorins (39), members of this plexin family could participate in HTLV-I: an axomyelinic degeneration of the central nervous system. J. Neuro-
pathol. Exp. Neurol. 56:403.
sCD100 signaling. The presence of plexin-B1 in neural precursors 20. Osame, M., K. Arimura, M. Nakagawa, F. Umehara, K. Usuku, and S. Ijichi.
and the capacity of anti-plexin-B1 Ab to antagonize the T cell- 1997. HTLV-I associated myelopathy (HAM): review and recent studies. Leu-
induced apoptosis of neural precursors may support this hypothe- kemia 11(Suppl. 3):63.
21. t Hart, B. A., H. P. Brok, S. Amor, and R. E. Bontrop. 2001. The major histo-
sis. Thus, sCD100 and SEMA-3A can exert a similar paralytic compatibility complex influences the ethiopathogenesis of MS-like disease in
effect on immune cell migration via an identical cell surface re- primates at multiple levels. Hum. Immunol. 62:1371.
ceptor, while their deleterious effects leading to neural cell alter- 22. Jacobson, S. 2002. Immunopathogenesis of human T cell lymphotropic virus type
I-associated neurologic disease. J. Infect. Dis. 186(Suppl. 2):S187.
ation and death are mediated by different receptors. 23. Cuzner, M. L., D. Gveric, C. Strand, A. J. Loughlin, L. Paemen, G. Opdenakker,
In conclusion, this original paradigm of activated T lympho- and J. Newcombe. 1996. The expression of tissue-type plasminogen activator,
matrix metalloproteases and endogenous inhibitors in the central nervous system
cytes/neural cell interaction and the presence of sCD100- and in multiple sclerosis: comparison of stages in lesion evolution. J. Neuropathol.
CD100-positive infiltrating T cells in CNS of patients suffering of Exp. Neurol. 55:1194.
neuroinflammatory demyelination point to the role of the immune 24. Yamano, Y., M. Nagai, M. Brennan, C. A. Mora, S. S. Soldan, U. Tomaru,
N. Takenouchi, S. Izumo, M. Osame, and S. Jacobson. 2002. Correlation of
semaphorin CD100/SEMA-4D in the cross talk between the im- human T-cell lymphotropic virus type 1 (HTLV-1) mRNA with proviral DNA
mune system and the CNS. Immune semaphorins could be crucial load, virus-specific CD8⫹ T cells, and disease severity in HTLV-1-associated
myelopathy (HAM/TSP). Blood 99:88.
in demyelinating diseases, including MS and TSP/HAM, in which 25. Grant, C., K. Barmak, T. Alefantis, J. Yao, S. Jacobson, and B. Wigdahl. 2002.
a causal relationship among inflammation, oligodendrocyte loss, Human T cell leukemia virus type I and neurological disease: events in bone
The Journal of Immunology 1255

marrow, peripheral blood, and central nervous system during normal immune 39. Tamagnone, L., S. Artigiani, H. Chen, Z. He, G. I. Ming, H. Song, A. Chedotal,
surveillance and neuroinflammation. J. Cell. Physiol. 190:133. M. L. Winberg, C. S. Goodman, M. Poo, et al. 1999. Plexins are a large family
26. Blakemore, W. F., and R. J. Franklin. 2000. Transplantation options for thera- of receptors for transmembrane, secreted, and GPI-anchored semaphorins in ver-
peutic central nervous system remyelination. Cell Transplant. 9:289. tebrates. Cell 99:71.
27. Chang, A., W. W. Tourtellotte, R. Rudick, and B. D. Trapp. 2002. Premyelinating 40. Giraudon, P., S. Buart, A. Bernard, N. Thomasset, and M. F. Belin. 1996. Ex-
oligodendrocytes in chronic lesions of multiple sclerosis. N. Engl. J. Med. tracellular matrix-remodeling metalloproteinases and infection of the central ner-
346:165. vous system with retrovirus human T-lymphotropic virus type I (HTLV-I). Prog.
28. Horner, P. J., A. E. Power, G. Kempermann, H. G. Kuhn, T. D. Palmer, Neurobiol. 49:169.
J. Winkler, L. J. Thal, and F. H. Gage. 2000. Proliferation and differentiation of 41. Umehara, F., Y. Okada, N. Fujimoto, M. Abe, S. Izumo, and M. Osame. 1998.
progenitor cells throughout the intact adult rat spinal cord. J. Neurosci. 20:2218. Expression of matrix metalloproteinases and tissue inhibitors of metalloprotein-
29. Narayanan, S., L. Fu, E. Pioro, N. De Stefano, D. L. Collins, G. S. Francis, ases in HTLV-I-associated myelopathy. J. Neuropathol. Exp. Neurol. 57:839.
J. P. Antel, P. M. Matthews, and D. L. Arnold. 1997. Imaging of axonal damage 42. Neuhaus, O., J. J. Archelos, and H. P. Hartung. 2003. Immunomodulation in
in multiple sclerosis: spatial distribution of magnetic resonance imaging lesions. multiple sclerosis: from immunosuppression to neuroprotection. Trends Pharma-
Ann. Neurol. 41:385. col. Sci. 24:131.
30. Howard, A. K., D. K. Li, and J. Oger. 2003. MRI contributes to the differentiation 43. Steinman, L. 2001. Multiple sclerosis: a two-stage disease. Nat. Immun. 2:762.
between MS and HTLV-I associated myelopathy in British Columbian coastal 44. Bjartmar, C., and B. D. Trapp. 2001. Axonal and neuronal degeneration in mul-
natives. Can. J. Neurol. Sci. 30:41. tiple sclerosis: mechanisms and functional consequences. Curr. Opin. Neurol.
31. Coleman, M. P., and V. H. Perry. 2002. Axon pathology in neurological disease: 14:271.
a neglected therapeutic target. Trends Neurosci. 25:532. 45. Zhang, J., S. Markovic-Plese, B. Lacet, J. Raus, H. L. Weiner, and D. A. Hafler.
32. De Winter, F., M. Oudega, A. J. Lankhorst, F. P. Hamers, B. Blits, 1994. Increased frequency of interleukin 2-responsive T cells specific for myelin
M. J. Ruitenberg, R. J. Pasterkamp, W. H. Gispen, and J. Verhaagen. 2002. basic protein and proteolipid protein in peripheral blood and cerebrospinal fluid
Injury-induced class 3 semaphorin expression in the rat spinal cord. Exp. Neurol. of patients with multiple sclerosis. J. Exp. Med. 179:973.
175:61.
46. Lassmann, H. 2002. Mechanisms of demyelination and tissue destruction in mul-
33. Zhang, S. C., B. Ge, and I. D. Duncan. 1999. Adult brain retains the potential to
tiple sclerosis. Clin. Neurol. Neurosurg. 104:168.
generate oligodendroglial progenitors with extensive myelination capacity. Proc.
47. Pasterkamp, R. J., and J. Verhaagen. 2001. Emerging roles for semaphorins in
Natl. Acad. Sci. USA 96:4089.
neural regeneration. Brain Res. Brain Res. Rev. 35:36.
34. Popovic, M., G. Lange-Wantzin, P. S. Sarin, D. Mann, and R. C. Gallo. 1983.
Transformation of human umbilical cord blood T cells by human T-cell leukemia/ 48. Levine, J. M., R. Reynolds, and J. W. Fawcett. 2001. The oligodendrocyte pre-
lymphoma virus. Proc. Natl. Acad. Sci. USA 80:5402. cursor cell in health and disease. Trends Neurosci. 24:39.
35. Giraudon, P., R. Szymocha, S. Buart, A. Bernard, L. Cartier, M. F. Belin, and 49. Leist, M., and M. Jaattela. 2001. Four deaths and a funeral: from caspases to
H. Akaoka. 2000. T lymphocytes activated by persistent viral infection differen- alternative mechanisms. Nat. Rev. Mol. Cell Biol. 2:589.
tially modify the expression of metalloproteinases and their endogenous inhibi- 50. Wolswijk, G. 2002. Oligodendrocyte precursor cells in the demyelinated multiple
tors, TIMPs, in human astrocytes: relevance to HTLV-I-induced neurological sclerosis spinal cord. Brain 125:338.
disease. J. Immunol. 164:2718. 51. Levin, M. C., M. Krichavsky, J. Berk, S. Foley, M. Rosenfeld, J. Dalmau,
36. Derrington, E. A., N. Dufay, B. B. Rudkin, and M. F. Belin. 1998. Human prim- G. Chang, J. B. Posner, and S. Jacobson. 1998. Neuronal molecular mimicry in
itive neuroectodermal tumor cells behave as multipotent neural precursors in immune-mediated neurologic disease. Ann. Neurol. 44:87.
response to FGF2. Oncogene 17:1663. 52. Shi, W., A. Kumanogoh, C. Watanabe, J. Uchida, X. Wang, T. Yasui,
37. Buzanska, L., N. Spassky, M. F. Belin, A. Giangrande, F. Guillemot, C. Klambt, K. Yukawa, M. Ikawa, M. Okabe, J. R. Parnes, et al. 2000. The class IV sema-
M. Labouesse, J. L. Thomas, K. Domanska-Janik, and B. Zalc. 2001. Human phorin CD100 plays nonredundant roles in the immune system: defective B and
medulloblastoma cell line DEV is a potent tool to screen for factors influencing T cell activation in CD100-deficient mice. Immunity 13:633.
differentiation of neural stem cells. J. Neurosci. Res. 65:17. 53. Kikutani, H., and A. Kumanogoh. 2003. Semaphorins in interactions between T
38. Szymocha, R., H. Akaoka, M. Dutuit, C. Malcus, M. Didier-Bazes, M. F. Belin, cells and antigen-presenting cells. Nat. Rev. Immunol. 3:159.
and P. Giraudon. 2000. Human T-cell lymphotropic virus type 1-infected T lym- 54. Versijpt, J., K. Van Laere, R. A. Dierckx, F. Dumont, P. P. De Deyn, G. Slegers,
phocytes impair catabolism and uptake of glutamate by astrocytes via Tax-1 and and J. Korf. 2003. Scintigraphic visualization of inflammation in neurodegenera-
tumor necrosis factor ␣. J. Virol. 74:6433. tive disorders. Nucl. Med. Commun. 24:209.

Vous aimerez peut-être aussi